1
|
Mani R, Benrashid S, Templeton MD, Druhan LJ, Seegers SL, Chakraborty S, Teague SE, Jaros SC, Yang HT, Foureau DM, Steuerwald NM, Pal D, Ghosh N, Copelan EA, Durden DL, Avalos BR, Park SI. TP53 upregulation via aurora kinase inhibition overcomes primary failure to venetoclax in BCL2-rearranged lymphomas. iScience 2025; 28:112584. [PMID: 40491476 PMCID: PMC12146551 DOI: 10.1016/j.isci.2025.112584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 12/04/2024] [Accepted: 03/29/2025] [Indexed: 06/11/2025] Open
Abstract
Bcl2 inhibition has excellent antitumor activity against hematologic malignancies. However, the clinical results in lymphomas harboring BCL2 gene rearrangements have been disappointing, and the mechanism of this intrinsic resistance remains unknown. Herein, we report that Bcl2 inhibition rapidly repressed p53 with poor response in BCL2-rearranged lymphoma cells. However, concurrent inhibition of aurora kinase (Aurk) overcame this primary resistance to Bcl2 inhibition by restoring the p53/p21 proapoptotic axis via a post-transcriptional increase in p53. Two independent BCL2-rearranged lymphoma murine models showed complete tumor regression in all animals treated with combined Bcl2/Aurk inhibition, whereas mice treated with single-agents demonstrated rapid progression. Transcriptome analysis confirmed that BCL2-rearranged lymphomas rapidly downregulated the p53 target CDKN1A (p21) in response to Bcl2 inhibition in vivo. However, concurrent inhibition of Aurk restored the TP53/CDKN1A pathway, sensitizing the tumors to Bcl2 inhibitor-mediated apoptosis. These data lay the groundwork for evaluation of this combination in the clinical setting.
Collapse
Affiliation(s)
- Rajeswaran Mani
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Samon Benrashid
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Margaret D. Templeton
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Lawrence J. Druhan
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Sara L. Seegers
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Supriya Chakraborty
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Sarah E. Teague
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Scott C. Jaros
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Hsih-Te Yang
- Center for Cancer Biostatistics, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - David M. Foureau
- Immune Monitoring Laboratory, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Nury M. Steuerwald
- Molecular Biology and Genomics Laboratory, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Dhananjaya Pal
- Molecular Targeted Therapeutics, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Nilanjan Ghosh
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Edward A. Copelan
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Donald L. Durden
- Molecular Targeted Therapeutics, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Belinda R. Avalos
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Steven I. Park
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|
2
|
Allen B, Bottomly D, Köhnke T, Wang A, Lin HY, Johnson K, Kenna I, Streltsova A, Martin E, Chen R, Savoy L, Long N, Ryabinin P, Kurtz SE, Eide CA, Carlos A, Kaempf A, Liu T, Tognon C, Searles R, Piehowski PD, Gosline SJC, Agarwal A, Chang BH, Barton M, Druker BJ, McWeeney SK, Majeti R, Tyner JW, Zhang H. A CEBPB/IL-1β/TNF-α feedback loop drives drug resistance to venetoclax and MDM2 inhibitors in monocytic leukemia. Blood 2025; 145:2488-2506. [PMID: 40009487 PMCID: PMC12163741 DOI: 10.1182/blood.2024028239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/28/2025] Open
Abstract
ABSTRAT MDM2 inhibitors are promising therapeutics for acute myeloid leukemia (AML) with wild-type TP53. Through an integrated analysis of functional genomic data from primary patient samples, we found that an MDM2 inhibitor, idasanutlin, like venetoclax, is ineffective against monocytic leukemia (French-American-British [FAB] subtype M4/M5). To dissect the underlying resistance mechanisms, we explored both intrinsic and extrinsic factors. We found that monocytic leukemia cells express elevated levels of CEBPB, which promote monocytic differentiation, suppress CASP3 and CASP6, and upregulate MCL1, BCL2A1, and the interleukin (IL-1)/tumor necrosis factor alpha (TNF-α)/NF-κB pathway members, thereby conferring drug resistance to a broad range of MDM2 inhibitors, BH3 mimetics, and venetoclax combinations. In addition, aberrant monocytes in M4/M5 leukemia produce elevated levels of IL-1 and TNF-α, which promote monocytic differentiation and upregulate inflammatory cytokines and receptors, thereby extrinsically protecting leukemia blasts from venetoclax and MDM2 inhibition. Interestingly, IL-1β and TNF-α only increase CEBPB levels and protect M4/M5 cells from these drugs but not M0/M1 leukemia cells. Treatment with venetoclax and idasanutlin induces compensatory upregulation of CEBPB and the IL-1/TNF-α/NF-κB pathway independent of the FAB subtype, indicating drug-induced compensatory protection mechanisms. The combination of venetoclax or idasanutlin with inhibitors that block the IL-1/TNF-α pathway demonstrates synergistic cytotoxicity in M4/M5 AML. As such, we uncovered a targetable positive feedback loop that involves CEBPB, IL-1/TNF-α, and monocyte differentiation in M4/M5 leukemia and promotes both intrinsic and extrinsic drug resistance and drug-induced protection against venetoclax and MDM2 inhibitors.
Collapse
MESH Headings
- Humans
- Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors
- Sulfonamides/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Drug Resistance, Neoplasm/drug effects
- Interleukin-1beta/metabolism
- Interleukin-1beta/genetics
- CCAAT-Enhancer-Binding Protein-beta/metabolism
- CCAAT-Enhancer-Binding Protein-beta/genetics
- Leukemia, Monocytic, Acute/drug therapy
- Leukemia, Monocytic, Acute/metabolism
- Leukemia, Monocytic, Acute/pathology
- Leukemia, Monocytic, Acute/genetics
- Signal Transduction/drug effects
- Cell Line, Tumor
- Antineoplastic Agents/pharmacology
- Monocytes
Collapse
Affiliation(s)
- Basil Allen
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Daniel Bottomly
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Thomas Köhnke
- Division of Hematology, Department of Medicine, Cancer Institute and Institute of Stem Cell Biology and Regenerative Medicine, Stanford, CA
| | - Anthony Wang
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Hsin-Yun Lin
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Kara Johnson
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Isabel Kenna
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Anastatia Streltsova
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Emma Martin
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Reid Chen
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Lindsey Savoy
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Nicola Long
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Peter Ryabinin
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Stephen E. Kurtz
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Christopher A. Eide
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Amy Carlos
- Massively Parallel Sequencing Shared Resource, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Andy Kaempf
- Biostatistics Shared Resource, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Tingting Liu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Cristina Tognon
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Robert Searles
- Massively Parallel Sequencing Shared Resource, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Paul D. Piehowski
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Sara J. C. Gosline
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Anupriya Agarwal
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Bill H. Chang
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Michelle Barton
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR
| | - Brian J. Druker
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Shannon K. McWeeney
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Ravindra Majeti
- Division of Hematology, Department of Medicine, Cancer Institute and Institute of Stem Cell Biology and Regenerative Medicine, Stanford, CA
| | - Jeffrey W. Tyner
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Haijiao Zhang
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| |
Collapse
|
3
|
Li Y, Wang F. Research Progress on Traditional Chinese Medicines Reversing Multidrug Resistance and Mechanisms in Lung Cancer. Cancer Biother Radiopharm 2025. [PMID: 40402865 DOI: 10.1089/cbr.2025.0078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2025] Open
Abstract
Lung cancer continues to be a primary contributor to cancer-related deaths globally, and multidrug resistance (MDR) poses a significant obstacle in its management. Traditional Chinese medicines (TCMs), recognized for their comprehensive therapeutic strategies and low incidence of adverse effects, have garnered attention due to their capacity to mitigate MDR in cancer cells. Nevertheless, deciphering the precise mechanisms through which TCMs reverse MDR in lung cancer presents a substantial scientific challenge. The objective of this review is to examine prevalent manifestations of MDR in lung cancer and underscore recent advancements in understanding how TCMs might surmount this form of resistance. The review begins by investigating the unique characteristics of TCMs and their pivotal function in reversing MDR in lung cancer. Subsequently, it explores various forms of MDR in lung cancer, such as aberrant expression of cell membrane transport proteins, dysregulation of intracellular enzyme systems, disrupted apoptosis, and heightened cellular repair mechanisms, emphasizing their detrimental impact on lung cancer treatment outcomes. Central to this review is a thorough analysis of the intricate mechanisms by which TCMs counteract MDR, along with an assessment of their efficacy in lung cancer therapy. Based on this analysis, the review offers insights into potential future research directions for utilizing TCMs to overcome MDR. This review seeks to provide a thorough examination of the role of TCMs in reversing MDR in lung cancer and to stimulate additional research into their clinical applications.
Collapse
Affiliation(s)
- Yuying Li
- School of Clinic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chengdu Shuangliu District Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Fei Wang
- School of Clinic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
Ryan P, Lee J. In vitro senescence and senolytic functional assays. Biomater Sci 2025. [PMID: 40375674 DOI: 10.1039/d4bm01684j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
A detailed understanding of aging biology and the development of anti-aging therapeutic strategies remain imperative yet inherently challenging due to the protracted nature of aging. Cellular senescence arises naturally through replicative exhaustion and is accelerated by clinical treatments or environmental stressors. The accumulation of senescent cells-defined by a loss of mitogenic potential, resistance to apoptosis, and acquisition of a pro-inflammatory secretory phenotype-has been implicated as a key driver of chronic disease, tissue degeneration, and organismal aging. Recent studies have highlighted the therapeutic promise of senolytic drugs, which selectively eliminate senescent cells. Compelling results from preclinical animal studies and ongoing clinical trials underscore this potential. However, the clinical translation of senolytics requires further pharmacological validation to refine selectivity, minimize toxicity, and determine optimal dosing. Equally important is the evaluation of senolytics' potential to restore tissue structure and function by reducing the senescent cell burden. In vitro tissue culture models offer a powerful platform to advance these efforts. This review summarizes the current landscape of in vitro systems used for inducing cellular senescence-referred to as "senescence assays"-and for screening senolytic drugs-referred to as "senolytic assays". We conclude by discussing key challenges to improving mechanistic insight, predictive accuracy, and clinical relevance in senolytic drug development, as well as emerging applications of senolytic therapies.
Collapse
Affiliation(s)
- Patrick Ryan
- Molecular & Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, 01003, USA.
| | - Jungwoo Lee
- Molecular & Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, 01003, USA.
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts, 01003, USA
| |
Collapse
|
5
|
Adam N, Yang Y, Djamshidi M, Seifan S, Ting NSY, Glover J, Touret N, Gordon PMK, Vineetha Warriyar KV, Krowicki H, Garcia CK, Savage SA, Goodarzi AA, Baird DM, Beattie TL, Riabowol K. hTERT Increases TRF2 to Induce Telomere Compaction and Extend Cell Replicative Lifespan. Aging Cell 2025:e70105. [PMID: 40371663 DOI: 10.1111/acel.70105] [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: 08/12/2024] [Revised: 04/22/2025] [Accepted: 04/29/2025] [Indexed: 05/16/2025] Open
Abstract
Replicative senescence occurs in response to shortened telomeres and is triggered by ATM and TP53-mediated DNA damage signaling that blocks replication. hTERT lengthens telomeres, which is thought to block damage signaling and the onset of senescence. We find that normal diploid fibroblasts expressing hTERT mutants unable to maintain telomere length do not initiate DNA damage signaling and continue to replicate, despite having telomeres shorter than senescent cells. The TRF1 and TRF2 DNA binding proteins of the shelterin complex stabilize telomeres, and we find that expression of different mutant hTERT proteins decreases levels of the Siah1 E3 ubiquitin ligase that targets TRF2 to the proteasome, by increasing levels of the CDC20 and FBXO5 E3 ligases that target Siah1. This restores the TRF2:TRF1 ratio to block the activation of ATM and subsequent activation of TP53 that is usually associated with DNA damage-induced senescence signaling. All hTERT variants reduce DNA damage signaling, and this occurs concomitantly with telomeres assuming a more compact, denser conformation than senescent cells as measured by super-resolution microscopy. This indicates that hTERT variants induce TRF2-mediated telomere compaction that is independent of telomere length, and it plays a dominant role in regulating the DNA damage signaling that induces senescence and blocks replication of human fibroblasts. These observations support the idea that very short telomeres often seen in cancer cells may fail to induce senescence due to selective stabilization of components of the shelterin complex, increasing telomere density, rather than maintaining telomere length via the reverse transcriptase activity of hTERT.
Collapse
Affiliation(s)
- Nancy Adam
- Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, Departments of Biochemistry & Molecular Biology and/or Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Yang Yang
- Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, Departments of Biochemistry & Molecular Biology and/or Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mahbod Djamshidi
- Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, Departments of Biochemistry & Molecular Biology and/or Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sara Seifan
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, UK
| | - Nicholas S Y Ting
- Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, Departments of Biochemistry & Molecular Biology and/or Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Joel Glover
- Live Cell Imaging Laboratory, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nicolas Touret
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Paul M K Gordon
- Centre for Health Genomics and Informatics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - K V Vineetha Warriyar
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Hokan Krowicki
- Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, Departments of Biochemistry & Molecular Biology and/or Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Sharon A Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Aaron A Goodarzi
- Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, Departments of Biochemistry & Molecular Biology and/or Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Duncan M Baird
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, UK
| | - Tara L Beattie
- Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, Departments of Biochemistry & Molecular Biology and/or Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Karl Riabowol
- Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, Departments of Biochemistry & Molecular Biology and/or Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
6
|
Gdowicz-Kłosok A, Krześniak M, Łasut-Szyszka B, Butkiewicz D, Rusin M. Antibacterial Activity of the p53 Tumor Suppressor Protein-How Strong Is the Evidence? Int J Mol Sci 2025; 26:4416. [PMID: 40362653 PMCID: PMC12072856 DOI: 10.3390/ijms26094416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
The p53 tumor suppressor is best known for controlling the cell cycle, apoptosis, DNA repair, and metabolism, but it also regulates immunity and is able to impede the live cycle of viruses. For this reason, these infectious agents encode proteins which inactivate p53. However, what is less known is that p53 can also be inactivated by human pathogenic bacteria. It is probably not due to collateral damage, but specific targeting, because p53 could interfere with their multiplication. The mechanisms of the antibacterial activity of p53 are poorly known. However, they can be inferred from the results of high-throughput studies, which have identified more than a thousand p53-activated genes. As it turns out, many of these genes code proteins which have proven or plausible antibacterial functions like the efficient detection of bacteria by pattern recognition receptors, the induction of pro-inflammatory pyroptosis, the recruitment of immune cells, direct bactericidal activity, and the presentation of bacterial metabolites to lymphocytes. Probably there are more antibacterial, p53-regulated functions which were overlooked because laboratory animals are kept in sterile conditions. In this review, we present the outlines of some intriguing antibacterial mechanisms of p53 which await further exploration. Definitely, this area of research deserves more attention, especially in light of the appearance of antibiotic-resistant bacterial strains.
Collapse
Affiliation(s)
| | | | | | | | - Marek Rusin
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-101 Gliwice, Poland; (A.G.-K.); (M.K.); (B.Ł.-S.); (D.B.)
| |
Collapse
|
7
|
Münick P, Strubel A, Balourdas DI, Funk JS, Mernberger M, Osterburg C, Dreier B, Schaefer JV, Tuppi M, Yüksel B, Schäfer B, Knapp S, Plückthun A, Stiewe T, Joerger AC, Dötsch V. DARPin-induced reactivation of p53 in HPV-positive cells. Nat Struct Mol Biol 2025; 32:790-801. [PMID: 39789211 DOI: 10.1038/s41594-024-01456-7] [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: 02/01/2024] [Accepted: 11/21/2024] [Indexed: 01/12/2025]
Abstract
Infection of cells with high-risk strains of the human papillomavirus (HPV) causes cancer in various types of epithelial tissue. HPV infections are responsible for ~4.5% of all cancers worldwide. Tumorigenesis is based on the inactivation of key cellular control mechanisms by the viral proteins E6 and E7. The HPV E6 protein interacts with the cellular E3 ligase E6AP, and this complex binds to the p53 DNA-binding domain, which results in degradation of p53. Inhibition of this interaction has the potential to reactivate p53, thus preventing oncogenic transformation. Here we describe the characterization of a designed ankyrin repeat protein that binds to the same site as the HPV E6 protein, thereby displacing the E3 ligase and stabilizing p53. Interaction with the designed ankyrin repeat protein does not affect p53 DNA binding or the crucial MDM2 negative feedback loop but reactivates a p53-dependent transcriptional program in HeLa (HPV18-positive) and SiHa (HPV16-positive) cells, suggesting a potential therapeutic use.
Collapse
Affiliation(s)
- Philipp Münick
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
| | - Alexander Strubel
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
| | - Dimitrios-Ilias Balourdas
- Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt, Germany
- Structural Genomics Consortium, Goethe University, Frankfurt, Germany
| | - Julianne S Funk
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Philipps-University, Marburg, Germany
| | - Marco Mernberger
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Philipps-University, Marburg, Germany
| | - Christian Osterburg
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
| | - Birgit Dreier
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Jonas V Schaefer
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Marcel Tuppi
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
| | - Büşra Yüksel
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
- IMPRS on Cellular Biophysics, Frankfurt, Germany
| | - Birgit Schäfer
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt, Germany
- Structural Genomics Consortium, Goethe University, Frankfurt, Germany
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Philipps-University, Marburg, Germany
- Genomics Core Facility, Philipps-University, Marburg, Germany
- Institute for Lung Health, Justus Liebig University, Giessen, Germany
| | - Andreas C Joerger
- Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt, Germany
- Structural Genomics Consortium, Goethe University, Frankfurt, Germany
| | - Volker Dötsch
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany.
| |
Collapse
|
8
|
Boutelle AM, Mabene AR, Yao D, Xu H, Wang M, Tang YJ, Lopez SS, Sinha S, Demeter J, Cheng R, Benard BA, McCrea EM, Valente LJ, Drainas AP, Fischer M, Majeti R, Petrov DA, Jackson PK, Yang F, Winslow MM, Bassik MC, Attardi LD. Integrative multiomic approaches reveal ZMAT3 and p21 as conserved hubs in the p53 tumor suppression network. Cell Death Differ 2025:10.1038/s41418-025-01513-8. [PMID: 40263541 DOI: 10.1038/s41418-025-01513-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 02/18/2025] [Accepted: 04/07/2025] [Indexed: 04/24/2025] Open
Abstract
TP53, the most frequently mutated gene in human cancer, encodes a transcriptional activator that induces myriad downstream target genes. Despite the importance of p53 in tumor suppression, the specific p53 target genes important for tumor suppression remain unclear. Recent studies have identified the p53-inducible gene Zmat3 as a critical effector of tumor suppression, but many questions remain regarding its p53-dependence, activity across contexts, and mechanism of tumor suppression alone and in cooperation with other p53-inducible genes. To address these questions, we used Tuba-seqUltra somatic genome editing and tumor barcoding in a mouse lung adenocarcinoma model, combinatorial in vivo CRISPR/Cas9 screens, meta-analyses of gene expression and Cancer Dependency Map data, and integrative RNA-sequencing and shotgun proteomic analyses. We established Zmat3 as a core component of p53-mediated tumor suppression and identified Cdkn1a as the most potent cooperating p53-induced gene in tumor suppression. We discovered that ZMAT3/CDKN1A serve as near-universal effectors of p53-mediated tumor suppression that regulate cell division, migration, and extracellular matrix organization. Accordingly, combined Zmat3-Cdkn1a inactivation dramatically enhanced cell proliferation and migration compared to controls, akin to p53 inactivation. Together, our findings place ZMAT3 and CDKN1A as hubs of a p53-induced gene program that opposes tumorigenesis across various cellular and genetic contexts.
Collapse
Affiliation(s)
- Anthony M Boutelle
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Aicha R Mabene
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - David Yao
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Haiqing Xu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Mengxiong Wang
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuning J Tang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Steven S Lopez
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Sauradeep Sinha
- Department of Bioengineering, Stanford University Schools of Engineering and Medicine, Stanford, CA, USA
| | - Janos Demeter
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ran Cheng
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Brooks A Benard
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Edel M McCrea
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Liz J Valente
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Heligenics Inc, Las Vegas, NV, USA
| | | | - Martin Fischer
- Computational Biology Group, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Ravindra Majeti
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Dmitri A Petrov
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Peter K Jackson
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Fan Yang
- Department of Bioengineering, Stanford University Schools of Engineering and Medicine, Stanford, CA, USA
| | - Monte M Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael C Bassik
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura D Attardi
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
9
|
Piergentili R, Sechi S. Targeting Regulatory Noncoding RNAs in Human Cancer: The State of the Art in Clinical Trials. Pharmaceutics 2025; 17:471. [PMID: 40284466 PMCID: PMC12030637 DOI: 10.3390/pharmaceutics17040471] [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: 01/22/2025] [Revised: 03/29/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025] Open
Abstract
Noncoding RNAs (ncRNAs) are a heterogeneous group of RNA molecules whose classification is mainly based on arbitrary criteria such as the molecule length, secondary structures, and cellular functions. A large fraction of these ncRNAs play a regulatory role regarding messenger RNAs (mRNAs) or other ncRNAs, creating an intracellular network of cross-interactions that allow the fine and complex regulation of gene expression. Altering the balance between these interactions may be sufficient to cause a transition from health to disease and vice versa. This leads to the possibility of intervening in these mechanisms to re-establish health in patients. The regulatory role of ncRNAs is associated with all cancer hallmarks, such as proliferation, apoptosis, invasion, metastasis, and genomic instability. Based on the function performed in carcinogenesis, ncRNAs may behave either as oncogenes or tumor suppressors. However, this distinction is not rigid; some ncRNAs can fall into both classes depending on the tissue considered or the target molecule. Furthermore, some of them are also involved in regulating the response to traditional cancer-therapeutic approaches. In general, the regulation of molecular mechanisms by ncRNAs is very complex and still largely unclear, but it has enormous potential both for the development of new therapies, especially in cases where traditional methods fail, and for their use as novel and more efficient biomarkers. Overall, this review will provide a brief overview of ncRNAs in human cancer biology, with a specific focus on describing the most recent ongoing clinical trials (CT) in which ncRNAs have been tested for their potential as therapeutic agents or evaluated as biomarkers.
Collapse
|
10
|
Mével-Aliset M, Radu AG, Allard J, Blanchet S, Montellier E, Hainaut P, Rossignol R, Torch S, Orsi GA, Thibert C. Transcriptional regulation by LKB1 in lung adenocarcinomas: Exploring oxidative stress, neuroglial and amino acid signatures. Biochem Biophys Res Commun 2025; 755:151571. [PMID: 40043609 DOI: 10.1016/j.bbrc.2025.151571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 02/19/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
Lung adenocarcinoma (LUAD) is one of the most prevalent cancer types worldwide and has one of the poorest survival rates. Understanding its developpment is crucial for improving diagnosis, prognosis, and treatment. A key factor in LUAD is the frequent loss-of-function mutations in LKB1/STK11, a kinase that regulates metabolism. These mutations are linked to increased metastasis and worse clinical outcomes. In this study, we analyzed gene expression data from LUAD patients to explore how LKB1 mutations affect cancer behavior. We found that LKB1 mutations in KRAS-driven LUAD lead to widespread gene downregulation. By integrating avalaible protein interaction data, mass spectrometry analysis of LKB1 nuclear partners, and co-immunoprecipitations experiments, we identified BRG1, a chromatin activator and subunit of the BAF complex, as a nuclear partner of LKB1. Further analysis suggested that LKB1 mutations may impair BRG1 activity, disrupting chromatin regulation and gene expression. Notably, LUAD patients with mutated LKB1 showed gene expression patterns indicative of oxidative stress, defective neuronal-glial and neuroinflammation programs, and altered amino acid homeostasis. These changes resemble the roles LKB1 plays in neural crest stem cells, suggesting that LKB1 may reduce tumor aggressiveness in LUAD by maintaining a developmental gene expression program.
Collapse
Affiliation(s)
- Marie Mével-Aliset
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Anca G Radu
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Jordan Allard
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Sandrine Blanchet
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Emilie Montellier
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Pierre Hainaut
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Rodrigue Rossignol
- INSERM U1211, Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France; CELLOMET, Functional Genomics Center (CGFB), 146 rue Léo Saignat, 33076, Bordeaux, France
| | - Sakina Torch
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Guillermo A Orsi
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics of Regeneration and Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Chantal Thibert
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France.
| |
Collapse
|
11
|
Agrawal R, Sengupta S. p53 regulates DREAM complex-mediated repression in a p21-independent manner. EMBO J 2025; 44:2279-2297. [PMID: 40038454 PMCID: PMC12000331 DOI: 10.1038/s44318-025-00402-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 01/18/2025] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
The DREAM repressor complex regulates genes involved in the cell cycle and DNA repair, vital for maintaining genome stability. Although it mediates p53-driven repression through the canonical p53-p21-Rb axis, the potential for p53 to directly regulate DREAM targets independently of its transcriptional activity has not been explored. Here, we demonstrate that in asynchronously growing cells, p53 loss leads to greater de-repression of DREAM targets compared to p21 loss alone. Both wild-type and transactivation-deficient p53 mutants are capable of repressing DREAM targets, suggesting a transactivation-independent "non-canonical" repression mechanism. These p53 variants bind p130/p107, irrespective of their phosphorylation status, while cancer-associated p53 mutants disrupt DREAM complex function by sequestering E2F4. Re-ChIP analysis shows co-recruitment of p53 and E2F4 to known and newly identified DREAM target promoters, indicating direct repression of these targets by p53. These findings reveal a novel, transactivation-independent mechanism of p53-mediated repression, expanding our understanding of p53's tumor-suppressive functions and suggesting DREAM complex targeting as potential future avenues in cancer therapy.
Collapse
Affiliation(s)
- Ritu Agrawal
- Biotechnology Research and Innovation Council-National Institute of Immunology (BRIC-NII), Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Sagar Sengupta
- Biotechnology Research and Innovation Council-National Institute of Immunology (BRIC-NII), Aruna Asaf Ali Marg, New Delhi, 110067, India.
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), PO: NSS, Kalyani, 741251, India.
| |
Collapse
|
12
|
Catalano A, Haas LS, Zodel K, Adlesic M, Cuomo F, Peighambari A, Metzger P, Huang H, Haug S, Köttgen A, Köhler N, Boerries M, Frew IJ. Mutations in tumor suppressor genes Vhl and Rassf1a cause DNA damage, chromosomal instability and induce gene expression changes characteristic of clear cell renal cell carcinoma. Kidney Int 2025; 107:666-686. [PMID: 39725222 DOI: 10.1016/j.kint.2024.12.003] [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: 01/29/2024] [Revised: 11/06/2024] [Accepted: 12/04/2024] [Indexed: 12/28/2024]
Abstract
RASSF1A is frequently biallelically inactivated in clear cell renal cell carcinoma (ccRCC) due to loss of chromosome 3p and promoter hypermethylation. Here we investigated the cellular and molecular consequences of single and combined deletion of the Rassf1a and Vhl tumor suppressor genes to model the common ccRCC genotype of combined loss of function of RASSF1A and VHL. In mouse embryonic fibroblasts and in primary kidney epithelial cells, double deletion of Rassf1a and Vhl caused chromosomal segregation defects and increased formation of micronuclei, demonstrating that pVHL and RASSF1A function to maintain genomic integrity. Combined Rassf1a and Vhl deletion in kidney epithelial cells in vivo increased proliferation and caused mild tubular disorganization, but did not lead to the development of kidney tumors. Single cell RNA-sequencing unexpectedly revealed that Rassf1a or Vhl deletion both induce the expression of an overlapping set of genes in a sub-population of proximal tubule cells. Many of these genes are also upregulated in the Vhl/Trp53/Rb1 deficient mouse model of ccRCC. In other subsets of proximal tubule cells, combined Vhl/Rassf1a deletion induced the expression of additional genes that were not upregulated in each of the single knockouts. The expression of the human homologues of Rassf1a-regulated genes correlate negatively with RASSF1 expression levels in human ccRCC. Our results suggest that the loss of RASSF1A function establishes a ccRCC-characteristic gene expression pattern.
Collapse
Affiliation(s)
- Antonella Catalano
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Laura S Haas
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kyra Zodel
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mojca Adlesic
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Francesca Cuomo
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Asin Peighambari
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Patrick Metzger
- Institute of Medical Bioinformatics and Systems Medicine, Medical Centre-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hsin Huang
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan Haug
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Natalie Köhler
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Centre-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Comprehensive Cancer Center Freiburg (CCCF), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Ian J Frew
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Comprehensive Cancer Center Freiburg (CCCF), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, Faculty of Biology University of Freiburg, Freiburg, Germany.
| |
Collapse
|
13
|
Kaatsch HL, Kubitscheck L, Wagner S, Hantke T, Preiss M, Ostheim P, Nestler T, Piechotka J, Overhoff D, Brockmann MA, Waldeck S, Port M, Ullmann R, Becker BV. Routine CT Diagnostics Cause Dose-Dependent Gene Expression Changes in Peripheral Blood Cells. Int J Mol Sci 2025; 26:3185. [PMID: 40243988 PMCID: PMC11989232 DOI: 10.3390/ijms26073185] [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: 03/11/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
The increasing use of computed tomography (CT) has led to a rise in cumulative radiation dose due to medical imaging, raising concerns about potential long-term adverse effects. Large-scale epidemiological studies indicate a higher tumor incidence associated with CT examinations, but the underlying biological mechanisms remain largely unexplained. To gain further insights into the cellular response triggered by routine CT diagnostics, we investigated CT-induced changes of gene expression in peripheral blood cells using whole transcriptome sequencing. RNA was isolated from peripheral blood cells of 40 male patients with asymptomatic microhematuria, sampled before and after multi-phase abdominal CT (CTDIvol: 3.75-26.95 mGy, median: 6.55 mGy). On average, 22.11 million sequence reads (SD 5.71) per sample were generated to identify differentially expressed genes 6 h post-exposure by means of DESeq2. To assess the dose dependency of CT-induced effects, we additionally divided samples into three categories: low exposure (≤6.55 mGy, n = 20), medium exposure (>6.55 mGy and <12 mGy, n = 16), and high exposure (≥12 mGy, n = 4), and repeated gene expression analysis for each subset and their corresponding prae-exposure sample. CT exposure caused consistent and dose-dependent upregulation of six genes (EDA2R, AEN, FDXR, DDB2, PHLDA3, and MIR34AHG; padj < 0.1). These genes share several functional commonalities, including regulation by TP53 and involvement in the DNA damage response. The biological pathways highlighted by Gene Set Enrichment Analysis (GSEA) suggest a dose-dependent increase of cellular damage and metabolic particularities in the low-exposure subset, which may be related to a potential adaptive cellular response to low-dose irradiation. Irrespective of applied dose, AEN emerged as the most robust biomarker for CT exposure among all genes. Routine abdominal CT scans cause dose-dependent gene deregulation in association with DNA damage in peripheral blood cells after in vivo exposure. Regarding risk assessment of CT, our results support the commonly applied "As Low-As -Reasonably Achievable (ALARA)" principle. Evidence of additional gene expression changes associated with metabolic processes indicates a rather complex molecular response beyond DNA damage after CT exposure, and emphasizes the need for further targeted investigations.
Collapse
Affiliation(s)
- Hanns Leonhard Kaatsch
- Department of Radiology and Neuroradiology, Bundeswehr Central Hospital, 56072 Koblenz, Germany; (H.L.K.)
- Bundeswehr Institute of Radiobiology affiliated to Ulm University, 80937 Munich, Germany
| | - Laura Kubitscheck
- Bundeswehr Institute of Radiobiology affiliated to Ulm University, 80937 Munich, Germany
| | - Simon Wagner
- Bundeswehr Institute of Radiobiology affiliated to Ulm University, 80937 Munich, Germany
| | - Thomas Hantke
- Bundeswehr Institute of Radiobiology affiliated to Ulm University, 80937 Munich, Germany
| | - Maximilian Preiss
- Bundeswehr Institute of Radiobiology affiliated to Ulm University, 80937 Munich, Germany
| | - Patrick Ostheim
- Bundeswehr Institute of Radiobiology affiliated to Ulm University, 80937 Munich, Germany
| | - Tim Nestler
- Department of Urology, Bundeswehr Central Hospital, 56072 Koblenz, Germany
| | - Joel Piechotka
- Department of Radiology and Neuroradiology, Bundeswehr Central Hospital, 56072 Koblenz, Germany; (H.L.K.)
| | - Daniel Overhoff
- Department of Radiology and Neuroradiology, Bundeswehr Central Hospital, 56072 Koblenz, Germany; (H.L.K.)
| | - Marc A. Brockmann
- Department of Neuroradiology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Stephan Waldeck
- Department of Radiology and Neuroradiology, Bundeswehr Central Hospital, 56072 Koblenz, Germany; (H.L.K.)
- Department of Neuroradiology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Matthias Port
- Bundeswehr Institute of Radiobiology affiliated to Ulm University, 80937 Munich, Germany
| | - Reinhard Ullmann
- Bundeswehr Institute of Radiobiology affiliated to Ulm University, 80937 Munich, Germany
| | - Benjamin V. Becker
- Department of Radiology and Neuroradiology, Bundeswehr Central Hospital, 56072 Koblenz, Germany; (H.L.K.)
- Bundeswehr Institute of Radiobiology affiliated to Ulm University, 80937 Munich, Germany
- Department of Neuroradiology, University Medical Center Mainz, 55131 Mainz, Germany
| |
Collapse
|
14
|
Ke X, van Soldt B, Vlahos L, Zhou Y, Qian J, George J, Capdevila C, Glass I, Yan K, Califano A, Cardoso WV. Morphogenesis and regeneration share a conserved core transition cell state program that controls lung epithelial cell fate. Dev Cell 2025; 60:819-836.e7. [PMID: 39667932 PMCID: PMC11945641 DOI: 10.1016/j.devcel.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 08/07/2024] [Accepted: 11/17/2024] [Indexed: 12/14/2024]
Abstract
Transitional cell states are at the crossroads of crucial developmental and regenerative events, yet little is known about how these states emerge and influence outcomes. The alveolar and airway epithelia arise from distal lung multipotent progenitors, which undergo cell fate transitions to form these distinct compartments. The identification and impact of cell states in the developing lung are poorly understood. Here, we identified a population of Icam1/Nkx2-1 epithelial progenitors harboring a transitional state program remarkably conserved in humans and mice during lung morphogenesis and regeneration. Lineage-tracing and functional analyses reveal their role as progenitors to both airways and alveolar cells and the requirement of this transitional program to make distal lung progenitors competent to undergo airway cell fate specification. The identification of a common progenitor cell state in vastly distinct processes suggests a unified program reiteratively regulating outcomes in development and regeneration.
Collapse
Affiliation(s)
- Xiangyi Ke
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pharmacology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Benjamin van Soldt
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lukas Vlahos
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Yizhuo Zhou
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Pulmonary & Allergy Critical Care, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jun Qian
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Joel George
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Digestive and Liver Disease, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Claudia Capdevila
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Digestive and Liver Disease, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ian Glass
- Birth Defects Research Laboratory (BDRL), University of Washington, Seattle, WA 98105, USA
| | - Kelley Yan
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Digestive and Liver Disease, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Andrea Califano
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Wellington V Cardoso
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Pulmonary & Allergy Critical Care, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
15
|
Li J, Zhang S, Wang B, Dai Y, Wu J, Liu D, Liang Y, Xiao S, Wang Z, Wu J, Zheng D, Chen X, Shi F, Tan K, Ding X, Song H, Zhang S, Lu M. Pharmacological rescue of mutant p53 triggers spontaneous tumor regression via immune responses. Cell Rep Med 2025; 6:101976. [PMID: 39986271 PMCID: PMC11970324 DOI: 10.1016/j.xcrm.2025.101976] [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: 06/12/2023] [Revised: 08/05/2024] [Accepted: 01/28/2025] [Indexed: 02/24/2025]
Abstract
Tumor suppressor p53 is the most frequently mutated protein in cancer, possessing untapped immune-modulating capabilities in anticancer treatment. Here, we investigate the efficacy and underlying mechanisms of pharmacological reactivation of mutant p53 in treating spontaneous tumors in mice. In the p53 R279W (equivalent to the human hotspot R282W) mouse model developing spontaneous tumors, arsenic trioxide (ATO) treatment through drinking water significantly prolongs the survival of mice, dependent on p53-R279W reactivation. Transient regressions of spontaneous T-lymphomas are observed in 70% of the ATO-treated mice, accompanied by interferon (IFN) response. In allograft models, the tumor-suppressive effect of reactivated p53-R279W is detectably reduced in both immunodeficient Rag1-/- and CD8+ T cell-depleted mice. ATO also activates the IFN pathway in human cancer cells harboring various p53 mutations, as well as in primary samples derived from the p53-mutant patient treated with ATO. Together, p53 could serve as an alternative therapeutic target for the development of immunotherapies.
Collapse
Affiliation(s)
- Jiabing Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shuang Zhang
- State Key Laboratory of Systems Medicine for Cancer, Institute for Personalized Medicine and Med-X, Institute School of Biomedical Engineering Research, Shanghai Jiao Tong University, Shanghai, China
| | - Baohui Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Yuting Dai
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiale Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Dianjia Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ying Liang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shujun Xiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhengyuan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiaqi Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Derun Zheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xueqin Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fangfang Shi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kai Tan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xianting Ding
- State Key Laboratory of Systems Medicine for Cancer, Institute for Personalized Medicine and Med-X, Institute School of Biomedical Engineering Research, Shanghai Jiao Tong University, Shanghai, China.
| | - Huaxin Song
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Sujiang Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Min Lu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
16
|
Haddad A, Golan-Lev T, Benvenisty N, Goldberg M. Genome-wide screening in human embryonic stem cells identifies genes and pathways involved in the p53 pathway. Mol Med 2025; 31:97. [PMID: 40082762 PMCID: PMC11907909 DOI: 10.1186/s10020-025-01141-5] [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: 09/09/2024] [Accepted: 02/25/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND The tumor suppressor protein, p53, which is mutated in half of human tumors, plays a critical role in cellular responses to DNA damage and maintenance of genome stability. Therefore, increasing our understanding of the p53 pathway is essential for improving cancer treatment and diagnosis. METHODS This study, which aimed to identify genes and pathways that mediate resistance to p53 upregulation, used genome-wide CRISPR-Cas9 loss-of-function screening done with Nutlin-3a, which inhibits p53-MDM2 interaction, resulting in p53 accumulation and apoptotic cell death. We used bioinformatics analysis for the identification of genes and pathways that are involved in the p53 pathway and cell survival assays to validate specific genes. In addition, we used RNA-seq to identify differentially expressed p53 target genes in gene knockout (KO) cell lines. RESULTS Our screen revealed three significantly enriched pathways: The heparan sulfate glycosaminoglycan biosynthesis, diphthamide biosynthesis and Hippo pathway. Notably, TRIP12 was significantly enriched in our screen. We found that TRIP12 is required for the p53-dependent transcription of several pro-apoptotic genes. CONCLUSION Our study has identified two novel pathways that play a role in p53-mediated growth restriction. Moreover, we have highlighted the interaction between the Hippo and the p53 pathways. Interestingly, we have shown that TRIP12 plays an important function in the p53 pathway by selectively affecting its role as a transcription factor.
Collapse
Affiliation(s)
- Amir Haddad
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, the Hebrew University of Jerusalem, 91904, Jerusalem, Israel
- The Azrieli Center for Stem Cells and Genetic Research, The Alexander Silberman Institute of Life Sciences, the Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Tamar Golan-Lev
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, the Hebrew University of Jerusalem, 91904, Jerusalem, Israel
- The Azrieli Center for Stem Cells and Genetic Research, The Alexander Silberman Institute of Life Sciences, the Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Nissim Benvenisty
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, the Hebrew University of Jerusalem, 91904, Jerusalem, Israel
- The Azrieli Center for Stem Cells and Genetic Research, The Alexander Silberman Institute of Life Sciences, the Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Michal Goldberg
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, the Hebrew University of Jerusalem, 91904, Jerusalem, Israel.
| |
Collapse
|
17
|
Kadam L, Chan K, Ahuna K, Marshall N, Myatt L. Differential activation of p53-Lamin A/C and p16-RB mediated senescence pathways in trophoblast from pregnancies complicated by type A2 Gestational Diabetes Mellitus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.04.641461. [PMID: 40093078 PMCID: PMC11908226 DOI: 10.1101/2025.03.04.641461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Gestational diabetes mellitus (GDM) increases maternal risks such as hypertension and future type 2 diabetes while also contributing to fetal complications such as large-for-gestational-age infants and stillbirth. The placenta which is crucial for fetal development, exhibits structural and functional changes in GDM, but the impact of these alterations on placental trophoblast function remains unclear. During their differentiation villous cytotrophoblast display several characteristics of senescent cells however the role of senescence pathways in placental function remains unexplored in GDM. Here we investigate whether placental senescence pathways are altered in GDM, utilizing term villous tissue and primary trophoblasts to assess molecular changes, and determined fetal sex-based differences. Our data suggest that both p21 and p16 mediated senescence pathways are activated during trophoblast differentiation and are dysregulated in GDM placenta in a sexually dimorphic manner. We also provide evidence for increased activation of p53-Lamin A/C and p16-RB pathways in trophoblast from GDM placentas. Reduced expression of p21 and its downstream effects on GCM1 expression and βhCG secretion outline how altered physiological senescence can affect trophoblast differentiation and function. This is a seminal study highlighting how placental senescence pathways are altered in pregnancies complicated by GDM.
Collapse
|
18
|
Miller KN, Li B, Pierce-Hoffman HR, Patel S, Lei X, Rajesh A, Teneche MG, Havas AP, Gandhi A, Macip CC, Lyu J, Victorelli SG, Woo SH, Lagnado AB, LaPorta MA, Liu T, Dasgupta N, Li S, Davis A, Korotkov A, Hultenius E, Gao Z, Altman Y, Porritt RA, Garcia G, Mogler C, Seluanov A, Gorbunova V, Kaech SM, Tian X, Dou Z, Chen C, Passos JF, Adams PD. p53 enhances DNA repair and suppresses cytoplasmic chromatin fragments and inflammation in senescent cells. Nat Commun 2025; 16:2229. [PMID: 40044657 PMCID: PMC11882782 DOI: 10.1038/s41467-025-57229-3] [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: 11/21/2024] [Accepted: 02/13/2025] [Indexed: 03/09/2025] Open
Abstract
Genomic instability and inflammation are distinct hallmarks of aging, but the connection between them is poorly understood. Here we report a mechanism directly linking genomic instability and inflammation in senescent cells through a mitochondria-regulated molecular circuit involving p53 and cytoplasmic chromatin fragments (CCF) that are enriched for DNA damage signaling marker γH2A.X. We show that p53 suppresses CCF accumulation and its downstream inflammatory phenotype. p53 activation suppresses CCF formation linked to enhanced DNA repair and genome integrity. Activation of p53 in aged mice by pharmacological inhibition of MDM2 reverses transcriptomic signatures of aging and age-associated accumulation of monocytes and macrophages in liver. Mitochondrial ablation in senescent cells suppresses CCF formation and activates p53 in an ATM-dependent manner, suggesting that mitochondria-dependent formation of γH2A.X + CCF dampens nuclear DNA damage signaling and p53 activity. These data provide evidence for a mitochondria-regulated p53 signaling circuit in senescent cells that controls DNA repair, genome integrity, and senescence- and age-associated inflammation, with relevance to therapeutic targeting of age-associated disease.
Collapse
Affiliation(s)
- Karl N Miller
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA.
| | - Brightany Li
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | | | - Shreeya Patel
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Xue Lei
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Adarsh Rajesh
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Marcos G Teneche
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Aaron P Havas
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Armin Gandhi
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Carolina Cano Macip
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Jun Lyu
- Laboratory of Biochemistry and Molecular Biology; National Cancer Institute; National Institutes of Health, Bethesda, MD, USA
| | - Stella G Victorelli
- Department of Physiology and Biomedical Engineering; Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging; Mayo Clinic, Rochester, MN, USA
| | - Seung-Hwa Woo
- Department of Physiology and Biomedical Engineering; Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging; Mayo Clinic, Rochester, MN, USA
| | - Anthony B Lagnado
- Department of Physiology and Biomedical Engineering; Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging; Mayo Clinic, Rochester, MN, USA
| | - Michael A LaPorta
- NOMIS Center for Immunobiology and Microbial Pathogenesis; Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Tianhui Liu
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Nirmalya Dasgupta
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
- Center for Cancer Therapy; La Jolla Institute of Immunology, La Jolla, CA, USA
| | - Sha Li
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Andrew Davis
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Anatoly Korotkov
- Departments of Biology and Medicine; University of Rochester, Rochester, NY, USA
| | - Erik Hultenius
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Zichen Gao
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Yoav Altman
- Shared Resources; NCI-designated Cancer Center; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Rebecca A Porritt
- Shared Resources; NCI-designated Cancer Center; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Guillermina Garcia
- Shared Resources; NCI-designated Cancer Center; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Carolin Mogler
- Institute of Pathology; School of Medicine and Health; Technical University Munich (TUM), Munich, Germany
| | - Andrei Seluanov
- Departments of Biology and Medicine; University of Rochester, Rochester, NY, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine; University of Rochester, Rochester, NY, USA
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis; Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Xiao Tian
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Zhixun Dou
- Center for Regenerative Medicine, Department of Medicine; Massachusetts General Research Institute, Boston, MA, USA
- Harvard Stem Cell Institute; Harvard University, Cambridge, MA, USA
| | - Chongyi Chen
- Laboratory of Biochemistry and Molecular Biology; National Cancer Institute; National Institutes of Health, Bethesda, MD, USA
| | - João F Passos
- Department of Physiology and Biomedical Engineering; Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging; Mayo Clinic, Rochester, MN, USA
| | - Peter D Adams
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA.
| |
Collapse
|
19
|
Schmidt ENC, Evert BO, Pregler BEF, Melhem A, Hsieh M, Raspe M, Strobel H, Roos J, Pietsch T, Schuss P, Fischer‐Posovszky P, Westhoff M, Hölzel M, Herrlinger U, Vatter H, Waha A, Schneider M, Potthoff A. Tonabersat enhances temozolomide-mediated cytotoxicity in glioblastoma by disrupting intercellular connectivity through connexin 43 inhibition. Mol Oncol 2025; 19:878-898. [PMID: 39680504 PMCID: PMC11887680 DOI: 10.1002/1878-0261.13786] [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: 10/09/2023] [Revised: 10/24/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Glioblastoma cells rely on connexin 43 (Cx43)-based gap junctions (GJs) for intercellular communication, enabling them to integrate into a widely branched malignant network. Although there are promising prospects for new targeted therapies, the lack of clinically feasible GJ inhibitors has impeded their adoption in clinical practice. In the present study, we investigated tonabersat (TO), a blood-brain-barrier-penetrating drug with GJ-inhibitory properties, in regard to its potential to disassemble intercellular connectivity in glioblastoma networks. Fluorescence-guided measurements of calcein cell-to-cell transfer were used to study functional intercellular connectivity. Specific DNA fragmentation rates of propidium iodide-stained nuclei were measured as a surrogate readout for cell death using flow cytometry. CRISPR/Cas9-mediated gene editing of Cx43 served as a validation tool of cellular effects related to Cx43 GJ inhibition. 3' mRNA sequencing was performed for molecular downstream analysis. We found that TO reduced intercellular GJ-mediated cytosolic traffic and yielded a significant reduction of tumor microtube (TM) length. TO-mediated inhibition of cellular tumor networks was accompanied by a synergistic effect for temozolomide-induced cell death. CRISPR/Cas9 Cx43-knockout revealed similar results, indicating that TO-mediated inhibitory effects rely on the inhibition of Cx43-based GJs. Gene set enrichment analyses found that GJ-mediated synergistic cytotoxic effects were linked to a significant upregulation of cell death signaling pathways. In conclusion, TO disrupts TM-based network connectivity via GJ inhibition and renders glioblastoma cells more susceptible to cytotoxic therapy. Given its previous use in clinical trials for migraine therapy, TO might harbor the potential of bridging the idea of a GJ-targeted therapeutic approach from bench to bedside.
Collapse
Affiliation(s)
- Elena N. C. Schmidt
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Brain Tumor Translational Research GroupUniversity Hospital BonnGermany
| | | | - Barbara E. F. Pregler
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Brain Tumor Translational Research GroupUniversity Hospital BonnGermany
| | - Ahmad Melhem
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Brain Tumor Translational Research GroupUniversity Hospital BonnGermany
| | - Meng‐Chun Hsieh
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Brain Tumor Translational Research GroupUniversity Hospital BonnGermany
| | - Markus Raspe
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Brain Tumor Translational Research GroupUniversity Hospital BonnGermany
| | - Hannah Strobel
- Department of Pediatrics and Adolescent MedicineUniversity Medical Center UlmGermany
| | - Julian Roos
- Department of Pediatrics and Adolescent MedicineUniversity Medical Center UlmGermany
| | | | - Patrick Schuss
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Present address:
Department of NeurosurgeryBG Klinikum Unfallkrankenhaus Berlin BGGermany
| | - Pamela Fischer‐Posovszky
- Department of Pediatrics and Adolescent MedicineUniversity Medical Center UlmGermany
- German Center for Child and Adolescent Health (DZKJ), partner site UlmGermany
| | - Mike‐Andrew Westhoff
- Department of Pediatrics and Adolescent MedicineUniversity Medical Center UlmGermany
| | - Michael Hölzel
- Institute of Experimental OncologyUniversity Hospital BonnGermany
| | - Ulrich Herrlinger
- Department of Neurooncology, Center for Neurology and Center of Integrated Oncology ABCDUniversity Hospital BonnGermany
| | - Hartmut Vatter
- Department of NeurosurgeryUniversity Hospital BonnGermany
| | - Andreas Waha
- Department of NeuropathologyUniversity Hospital BonnGermany
| | - Matthias Schneider
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Brain Tumor Translational Research GroupUniversity Hospital BonnGermany
| | - Anna‐Laura Potthoff
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Brain Tumor Translational Research GroupUniversity Hospital BonnGermany
- Department of NeuropathologyUniversity Hospital BonnGermany
| |
Collapse
|
20
|
Kobar K, Tuzi L, Fiene JA, Burnley E, Galpin KJC, Midgen C, Laverty B, Subasri V, Wen TT, Hirst M, Moksa M, Carles A, Cao Q, Shlien A, Malkin D, Prykhozhij SV, Berman JN. tp53 R217H and R242H mutant zebrafish exhibit dysfunctional p53 hallmarks and recapitulate Li-Fraumeni syndrome phenotypes. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167612. [PMID: 39643218 DOI: 10.1016/j.bbadis.2024.167612] [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: 06/04/2024] [Revised: 10/28/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
Li-Fraumeni syndrome (LFS) is a hereditary cancer predisposition syndrome associated with a highly penetrant cancer spectrum characterized by germline TP53 mutations. We characterized the first LFS zebrafish hotspot mutants, tp53 R217H and R242H (human R248H and R273H), and found these mutants exhibit partial-to-no activation of p53 target genes, have defective cell-cycle checkpoints, and display partial-to-full resistance to apoptosis, although the R217H mutation has hypomorphic characteristics. Spontaneous tumor development histologically resembling human sarcomas was observed as early as 6 months. tp53 R242H mutants had a higher lifetime tumor incidence compared to tp53 null and R217H mutants, suggesting it is a more aggressive mutation. We observed mutation-specific tumor phenotypes across tp53 mutants with associated diverse transcriptomic and DNA methylome profiles in tp53 mutant larvae, impacting metabolism, cell signalling, and biomacromolecule synthesis and degradation. These tp53 zebrafish mutants demonstrate fidelity to their human counterparts and provide new insights into underlying tumorigenesis mechanisms and kinetics that suggest metabolic rewiring and cellular signalling changes occur prior to tumor initiation, which will guide targeted therapeutics for LFS.
Collapse
Affiliation(s)
- Kim Kobar
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Lissandra Tuzi
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jennifer A Fiene
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Erin Burnley
- Translational and Molecular Medicine Program, University of Ottawa, Ottawa, ON, Canada
| | | | - Craig Midgen
- Department of Pathology, IWK Health Centre, Halifax, NS, Canada
| | - Brianne Laverty
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Vallijah Subasri
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, ON, Canada; Peter Munk Cardiac Center, University Health Network, Toronto, ON, Canada
| | - Timmy T Wen
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, ON, Canada; Laboratory of Medicine and Pathobiology, University of Toronto, Canada
| | - Martin Hirst
- Department of Microbiology and Immunology, Michael Smith Laboratories, UBC, Vancouver, Canada; Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, Canada
| | - Michelle Moksa
- Department of Microbiology and Immunology, Michael Smith Laboratories, UBC, Vancouver, Canada
| | - Annaick Carles
- Department of Microbiology and Immunology, Michael Smith Laboratories, UBC, Vancouver, Canada
| | - Qi Cao
- Department of Microbiology and Immunology, Michael Smith Laboratories, UBC, Vancouver, Canada
| | - Adam Shlien
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, ON, Canada; Laboratory of Medicine and Pathobiology, University of Toronto, Canada
| | - David Malkin
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Department of Pediatrics, Division of Hematology/Oncology, Hospital for Sick Children, Toronto, ON, Canada
| | - Sergey V Prykhozhij
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Jason N Berman
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada; Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
21
|
Haidurov A, Zheltukhin AO, Snezhkina AV, Krasnov GS, Kudryavtseva AV, Budanov AV. p53-regulated SESN1 and SESN2 regulate cell proliferation and cell death through control of STAT3. Cell Commun Signal 2025; 23:105. [PMID: 39985075 PMCID: PMC11846189 DOI: 10.1186/s12964-025-02104-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 02/11/2025] [Indexed: 02/24/2025] Open
Abstract
Sestrin1 and Sestrin2 (SESN1&2) are evolutionarily conserved, stress-responsive proteins that regulate cell growth and viability. The primary target of Sestrins is the mTORC1 protein kinase, an activator of anabolic processes and an autophagy inhibitor. Our previous studies showed that inactivating SESN1&2 in lung adenocarcinoma A549 cells accelerates cell proliferation and confers resistance to cell death without affecting mTORC1 activity, suggesting that SESN1&2 modulate cellular processes via mTORC1-independent mechanisms. This work describes a new mechanism through which SESN1&2 regulate cell proliferation and death by suppressing the STAT3 transcription factor. Normally activated in response to stress and inflammation, STAT3 is frequently overactivated in human cancers. This overactivation promotes the expression of pro-proliferative and anti-apoptotic genes that drive carcinogenesis. We demonstrate that SESN1&2 inactivation stimulates STAT3 by downregulating the PTPRD phosphatase, a protein responsible for STAT3 dephosphorylation. Our study demonstrates that SESN1&2 deficiency may cause STAT3 activation and facilitate carcinogenesis and drug resistance, making SESN1&2 reactivation a potential cancer treatment strategy.
Collapse
Affiliation(s)
- Alexander Haidurov
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland
| | - Andrei O Zheltukhin
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia
| | - Anastasiya V Snezhkina
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia
| | - Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia
| | - Andrei V Budanov
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
22
|
Ribeiro JH, Etlioglu E, Buset J, Janssen A, Puype H, Berden L, Mbouombouo Mfossa AC, De Vos WH, Vermeirssen V, Baatout S, Rajan N, Quintens R. A human-specific, concerted repression of microcephaly genes contributes to radiation-induced growth defects in cortical organoids. iScience 2025; 28:111853. [PMID: 39967878 PMCID: PMC11834077 DOI: 10.1016/j.isci.2025.111853] [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: 07/10/2024] [Revised: 11/22/2024] [Accepted: 01/16/2025] [Indexed: 02/20/2025] Open
Abstract
Prenatal radiation-induced DNA damage poses a significant threat to neurodevelopment, resulting in microcephaly which primarily affects the cerebral cortex. So far, mechanistic studies were done in rodents. Here, we leveraged human cortical organoids to model fetal corticogenesis. Organoids were X-irradiated with moderate or high doses at different time points. Irradiation caused a dose- and time-dependent reduction in organoid size, which was more prominent in younger organoids. This coincided with a delayed and attenuated DNA damage response (DDR) in older organoids. Besides the DDR, radiation induced premature differentiation of neural progenitor cells (NPCs). Our transcriptomic analysis demonstrated a concerted p53-E2F4/DREAM-dependent repression of primary microcephaly genes, which was independently confirmed in cultured human NPCs and neurons. This was a human-specific feature, as it was not observed in mouse embryonic brains or primary NPCs. Thus, human cortical organoids are an excellent model for DNA damage-induced microcephaly and to uncover potentially targetable human-specific pathways.
Collapse
Affiliation(s)
- Jessica Honorato Ribeiro
- Radiobiology Unit, Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
- Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Emre Etlioglu
- Radiobiology Unit, Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
| | - Jasmine Buset
- Radiobiology Unit, Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
| | - Ann Janssen
- Radiobiology Unit, Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
| | - Hanne Puype
- Laboratory for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Lisa Berden
- Radiobiology Unit, Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
- Laboratory for Neurophysiology, Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
| | | | - Winnok H. De Vos
- Laboratory of Cell Biology and Histology, Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, 2610 Wilrijk, Belgium
| | - Vanessa Vermeirssen
- Laboratory for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
- Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Nicholas Rajan
- Radiobiology Unit, Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
| | - Roel Quintens
- Radiobiology Unit, Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
| |
Collapse
|
23
|
Sloan N, Mares J, Daly A, Coie L, Grier S, Barretto N, Casel O, Kang K, Jackson C, Pedersen M, Khiste S, Fullerton B, Petrescu J, Mattison C, Smith C, Suh Y, Menon V, Phatnani H. Uncovering the Signatures of Cellular Senescence in the Human Dorsolateral Prefrontal Cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.639091. [PMID: 40027780 PMCID: PMC11870546 DOI: 10.1101/2025.02.19.639091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Identifying senescent cells poses challenges due to their rarity, heterogeneity, and lack of a definitive marker. We performed Visium spatial transcriptomics (ST) and single nucleus RNA sequencing (snRNA-seq) on non-pathological human tissue to build a transcriptomic atlas of aging and senescence in the dorsolateral prefrontal cortex (dlPFC). We identified markers characteristic of aging dlPFC cortical layers and cell types. We also observed an increase in astrocyte abundance and decrease in somatostatin expressing inhibitory neurons. Overall, the senescence profile in the dlPFC was highly heterogeneous and heavily influenced by cell type identity and cortical layer. Combined unbiased analysis of ST and snRNA-seq datasets revealed gene expression modules encoding for communities of microglia and endothelial cells in the white matter and regional astrocytes programs that were strongly enriched with age and for senescence-related genes. These findings will help facilitate future studies exploring the function of senescent cell subpopulations in the aging brain.
Collapse
|
24
|
Rendo V, Lee EQ, Bossi C, Khuu N, Rudek MA, Pal S, Azazmeh N, Rashid R, Lin JR, Cusick M, Reynolds ARN, Fassinou ACR, Ayoub G, Malinowski S, Lapinskas E, Pisano W, Jeang J, Stopka SA, Regan MS, Spetz J, Desai A, Lieberman F, Palanichamy K, Fisher JD, Pelton K, Huang RY, Sarosiek KA, Nabors LB, Holdhoff M, Danda N, Strowd R, Desideri S, Walbert T, Ye X, Chakravarti A, Sorger PK, Santagata S, Agar NYR, Grossman SA, Alexander BM, Wen PY, Ligon KL, Beroukhim R. A window-of-opportunity trial reveals mechanisms of response and resistance to navtemadlin in patients with recurrent glioblastoma. Sci Transl Med 2025; 17:eadn6274. [PMID: 39970230 PMCID: PMC11994224 DOI: 10.1126/scitranslmed.adn6274] [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: 12/20/2023] [Revised: 09/16/2024] [Accepted: 01/29/2025] [Indexed: 02/21/2025]
Abstract
Inhibitors of murine double minute homolog 2 (MDM2) represent a promising therapeutic approach for the treatment of TP53 wild-type glioblastomas (GBMs), reactivating p53 signaling to induce cancer cell death. We conducted a surgical window-of-opportunity trial (NCT03107780) of the MDM2 inhibitor navtemadlin (KRT-232) in 21 patients with TP53 wild-type recurrent GBM to determine achievable drug concentrations within tumor tissues and biological mechanisms of response and resistance. Participants received navtemadlin at 120 mg (n = 10) or 240 mg (n = 11) for 2 days before surgical resection and after surgery until progression or unacceptable toxicity. Both 120 and 240 mg daily dosing achieved a pharmacodynamic impact, but median progression-free survival was 3.1 months. DNA sequencing of three recurrent tumors revealed an absence of TP53-inactivating mutations, indicating alternative mechanisms of resistance. To understand the mechanisms of response and resistance associated with navtemadlin, we conducted functional and spatial analyses of human tissue and patient-derived GBM neurosphere models. Navtemadlin induced partial tumor cell death as monotherapy, and combination with temozolomide enhanced apoptosis in GBM neurospheres while sparing normal bone marrow cells in vitro. We also observed up-regulation of oligodendrocyte differentiation genes with navtemadlin treatment and enrichment of oligodendrocyte transcription factor 2 (OLIG2)-positive cells at relapse, suggesting an unexplored mechanism of navtemadlin tolerance in GBM. Overall, these results indicated that clinically achievable doses of navtemadlin exert pharmacodynamic effects on GBM and suggest that combined treatment with temozolomide may be a route to more durable survival benefits.
Collapse
Affiliation(s)
- Veronica Rendo
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 05 Uppsala, Sweden
| | - Eudocia Q Lee
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Connor Bossi
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Nicholas Khuu
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michelle A Rudek
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Sangita Pal
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Narmen Azazmeh
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Rumana Rashid
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206-3701, USA
| | - Jia-Ren Lin
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Margaret Cusick
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Abigail R N Reynolds
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Auriole C R Fassinou
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Georges Ayoub
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Seth Malinowski
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Emily Lapinskas
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - William Pisano
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - John Jeang
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sylwia A Stopka
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michael S Regan
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Johan Spetz
- Department of Medical Radiation Sciences, Sahlgrenska Cancer Center, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Arati Desai
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Frank Lieberman
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Joy D Fisher
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Kristine Pelton
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Raymond Y Huang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kristopher A Sarosiek
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- John B. Little Center for Radiation Sciences, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Louis B Nabors
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Matthias Holdhoff
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Neeraja Danda
- Division of Hematology, Departments of Medicine and Oncology, Montefiore Medical Center, New York, NY 10467, USA
| | - Roy Strowd
- Department of Neurology, Wake Forest University, Winston-Salem, NC 27157, USA
| | - Serena Desideri
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Tobias Walbert
- Department of Neurology and Neurosurgery Henry Ford Health, Department of Neurology, Wayne State University and Michigan State University, Detroit, MI 48201, USA
| | - Xiaobu Ye
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Arnab Chakravarti
- Department of Radiation Oncology, Ohio State University College of Medicine, Columbus, OH 43210-1228, USA
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Sandro Santagata
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Nathalie Y R Agar
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Stuart A Grossman
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Brian M Alexander
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Patrick Y Wen
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Keith L Ligon
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Rameen Beroukhim
- Department of Medical Oncology and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
25
|
Miciak JJ, Petrova L, Sajwan R, Pandya A, Deckard M, Munoz AJ, Bunz F. Robust p53 phenotypes and prospective downstream targets in telomerase-immortalized human cells. Oncotarget 2025; 16:79-100. [PMID: 39969205 PMCID: PMC11837864 DOI: 10.18632/oncotarget.28690] [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: 10/31/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Cancers that retain wild type TP53 presumably harbor other clonal alterations that permitted their precursors to bypass p53-mediated growth suppression. Consequently, studies that employ TP53-wild type cancer cells and their isogenic derivatives may systematically fail to appreciate the full scope of p53 functionality. Several TP53 phenotypes are known to be absent in the widely used isogenic HCT116 colorectal cancer (CRC) model, which originated from a tumor that had retained wild type TP53. In contrast, we show that restoration of p53 in the TP53-mutant CRC cell line DLD-1 impeded cell proliferation, increased levels of senescence and sensitized cells to ionizing radiation (IR). To study p53 in a non-cancer context, we disrupted TP53 in hTERT-RPE1 cells. Derived from primary cells that were immortalized in vitro, hTERT-RPE1 expressed striking p53-dependent phenotypes and appeared to select for p53 loss during routine culture. hTERT-RPE1 expressed a p53-responsive transcriptome that was highly representative of diverse experimental systems. We discovered several novel downstream p53 targets of potential clinical relevance including ALDH3A1, which is involved in the detoxification of aldehydes and the metabolism of reactive oxygen species, and nectin cell adhesion molecule 4 (NECTIN4) which encodes a secreted surface protein that is overexpressed in many tumors.
Collapse
Affiliation(s)
- Jessica J. Miciak
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA
- Cellular and Molecular Medicine Graduate Program, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- These authors contributed equally to this work
| | - Lucy Petrova
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA
- These authors contributed equally to this work
| | - Rhythm Sajwan
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA
| | - Aditya Pandya
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA
| | - Mikayla Deckard
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA
| | - Andrew J. Munoz
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA
| | - Fred Bunz
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA
- Cellular and Molecular Medicine Graduate Program, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
26
|
Ji L, Shi Q, Shangguan Y, Chen C, Zhu J, Dong Z, Hong X, Liu X, Wei C, Zhu X, Li W. Molecular Response and Metabolic Reprogramming of the Spleen Coping with Cold Stress in the Chinese Soft-Shelled Turtle ( Pelodiscus sinensis). Antioxidants (Basel) 2025; 14:217. [PMID: 40002403 PMCID: PMC11852077 DOI: 10.3390/antiox14020217] [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: 01/08/2025] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
The Chinese soft-shelled turtle (Pelodiscus sinensis), as a type of warm-water reptile, could be induced to massive death by sharp temperature decline. Hence, the mechanism of spleen tissue responding to cold stress in the P. sinensis was investigated. The present results showed that the superoxide dismutase (SOD) activity declined from 4 to 16 days post-cold-stress (dps), while the catalase (CAT) and glutathione peroxidase (GSH-Px) activities increased, from 4 to 8 dps in the 14 °C (T14) and 7 °C (T7) stress groups. The spleen transcriptome in the T7 group and the control group (CG) at 4 dps obtained 2625 differentially expressed genes (DEGs), including 1462 upregulated and 1663 downregulated genes. The DEGs were enriched mainly in the pathways "intestinal immune network for IgA production" (Pigr, Il15ra, Tnfrsf17, Aicda, and Cd28), "toll-like receptor signaling pathway" (Mapk10, Tlr2, Tlr5, Tlr7, and Tlr8), and "cytokine-cytokine receptor interaction" (Cx3cl1, Cx3cr1, Cxcl14, Cxcr3, and Cxcr4). The metabolomic data showed that esculentic acid, tyrosol, diosgenin, heptadecanoic acid, and 7-ketodeoxycholic acid were obviously increased, while baccatin III, taurohyocholate, parthenolide, enterolactone, and tricin were decreased, in the CG vs. T7 comparison. Integrated analysis of the two omics revealed that "glycine, serine and threonine metabolism", "FoxO signaling pathway", and "neuroactive ligand-receptor interaction" were the main pathways responding to the cold stress. Overall, this work found that low temperature remarkably influenced the antioxidant enzyme activities, gene expression pattern, and metabolite profile in the spleen, indicating that immunity might be weakened by cold stress in P. sinensis.
Collapse
Affiliation(s)
- Liqin Ji
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510275, China; (L.J.); (X.L.)
| | - Qing Shi
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510275, China; (L.J.); (X.L.)
| | - Yisen Shangguan
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510275, China; (L.J.); (X.L.)
| | - Chen Chen
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510275, China; (L.J.); (X.L.)
| | - Junxian Zhu
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510275, China; (L.J.); (X.L.)
| | - Zhen Dong
- South China Sea Marine Survey Center, Key Laboratory of Marine Environmental Survey Technology and Application, Ministry of Natural Resources, Guangzhou 510275, China
| | - Xiaoyou Hong
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510275, China; (L.J.); (X.L.)
| | - Xiaoli Liu
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510275, China; (L.J.); (X.L.)
| | - Chengqing Wei
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510275, China; (L.J.); (X.L.)
| | - Xinping Zhu
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510275, China; (L.J.); (X.L.)
| | - Wei Li
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510275, China; (L.J.); (X.L.)
| |
Collapse
|
27
|
Berezovsky A, Nuga O, Datta I, Bergman K, Sabedot T, Gurdziel K, Irtenkauf S, Hasselbach L, Meng Y, Mueller C, . Petricoin EF, Brown S, Purandare N, Aras S, Mikkelsen T, Poisson L, Noushmehr H, Ruden D, deCarvalho AC. Impact of developmental state, p53 status, and interferon signaling on glioblastoma cell response to radiation and temozolomide treatment. PLoS One 2025; 20:e0315171. [PMID: 39919036 PMCID: PMC11805374 DOI: 10.1371/journal.pone.0315171] [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: 04/02/2024] [Accepted: 11/21/2024] [Indexed: 02/09/2025] Open
Abstract
Glioblastoma (GBM) tumors exhibit extensive genomic, epigenomic, and transcriptional diversity, with significant intratumoral heterogeneity, complicating standard treatment approaches involving radiation (RT) and the DNA-alkylating agent temozolomide (TMZ). In this study, we employed an integrative multi-omics approach, including targeted proteomics, transcriptomics, genomics, and DNA methylation profiling, to investigate the response of a representative panel of GBM patient-derived cancer stem cells (CSCs) to astrocytic differentiation and RT and TMZ treatments. Differentiated CSC progenies retained the expression of key stemness genes and survival pathways, while activating the BMP-Smad signaling pathway and upregulating extracellular matrix components. This was associated with increased resistance to TMZ, though not to RT, across all models. We identified TP53 status as a critical determinant of transcriptional response to both RT and TMZ, which was also modulated by the differentiation state and treatment modality in wildtype (wt) p53 GBM cells. Both mutant and wt p53 models exhibited significant activation of the DNA-damage associated interferon (IFN) response in CSCs and differentiated cells, implicating this pathway in the GBM response to therapy. We observed that activation of NF-κB was positively correlated with the levels of O-6-methylguanine-DNA methyltransferase (MGMT) protein, a direct DNA repair enzyme leading to TMZ resistance, regardless of MGMT promoter methylation status, further supporting the clinical potential for inhibition of NF-kB signaling in GBM treatment. Our integrative analysis of the impact of GBM cell developmental states, in the context of genomic and molecular diversity of patient-derived models, provides valuable insights for pre-clinical studies aimed at optimizing treatment strategies.
Collapse
Affiliation(s)
- Artem Berezovsky
- Department of Neurosurgery, Henry Ford Health, Detroit, Michigan, United States of America
- Department of Oncology, Wayne State University, Detroit, Michigan, United States of America
| | - Oluwademilade Nuga
- Department of Neurosurgery, Henry Ford Health, Detroit, Michigan, United States of America
- Department of Pharmacology, Wayne State University, Detroit, Michigan, United States of America
| | - Indrani Datta
- Department of Neurosurgery, Henry Ford Health, Detroit, Michigan, United States of America
| | - Kimberly Bergman
- Department of Neurosurgery, Henry Ford Health, Detroit, Michigan, United States of America
| | - Thais Sabedot
- Department of Neurosurgery, Henry Ford Health, Detroit, Michigan, United States of America
| | - Katherine Gurdziel
- Department of Pharmacology, Wayne State University, Detroit, Michigan, United States of America
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan, United States of America
| | - Susan Irtenkauf
- Department of Neurosurgery, Henry Ford Health, Detroit, Michigan, United States of America
| | - Laura Hasselbach
- Department of Neurosurgery, Henry Ford Health, Detroit, Michigan, United States of America
| | - Yuling Meng
- Department of Neurosurgery, Henry Ford Health, Detroit, Michigan, United States of America
| | - Claudius Mueller
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia, United States of America
| | - Emanuel F. . Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia, United States of America
| | - Stephen Brown
- Department of Radiation Oncology, Henry Ford Health, Detroit, Michigan, United States of America
| | - Neeraja Purandare
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan, United States of America
| | - Sidhesh Aras
- Department of Oncology, Wayne State University, Detroit, Michigan, United States of America
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan, United States of America
| | - Tom Mikkelsen
- Precision Medicine Program, Henry Ford Health, Detroit, Michigan, United States of America
| | - Laila Poisson
- Department of Public Health, Henry Ford Health, Detroit, Michigan, United States of America
| | - Houtan Noushmehr
- Department of Neurosurgery, Henry Ford Health, Detroit, Michigan, United States of America
| | - Douglas Ruden
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, Michigan, United States of America
| | - Ana C. deCarvalho
- Department of Neurosurgery, Henry Ford Health, Detroit, Michigan, United States of America
- Department of Oncology, Wayne State University, Detroit, Michigan, United States of America
- Department of Pharmacology, Wayne State University, Detroit, Michigan, United States of America
- Department of Physiology, College of Human Medicine, Michigan State University, East Lansing, Michigan, United States of America
| |
Collapse
|
28
|
Liu T, Li L, Yang Y, Li J, Yang X, Li L, Zheng Z, Yang B, Zhang P, Liu H. Effects of chronic cold stress and thermal stress on growth performance, hepatic apoptosis, oxidative stress, immune response and gut microbiota of juvenile hybrid sturgeon (Acipenser baerii ♀ × A. schrenkii ♂). FISH & SHELLFISH IMMUNOLOGY 2025; 157:110078. [PMID: 39642947 DOI: 10.1016/j.fsi.2024.110078] [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: 07/31/2024] [Revised: 11/10/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
The current study was conducted to investigate the effects of chronic cold stress and thermal stress on the growth performance, hepatic oxidative status, immune response, apoptosis and gut microbiota in juvenile hybrid sturgeon. The fish (initial mean weight: 21.4 ± 0.3 g) was reared at three temperatures (14 °C, 22 °C, and 30 °C) for 16 d, which were termed as low temperature group (LT), moderate temperature group (MT), and high temperature group (HT), respectively, and the second group was regarded as control group in this study. Each group was assigned randomly to three tanks with 15 fish per replica. The results indicated that cold stress resulted in a significant reduction of growth metrics and a significant increase of feed conversion ratio in fish compared with MT group. Interestingly, cold stress increased hepatocyte apoptosis revealed by TUNEL staining, along with nuclear disappearance in H&E-stained sections and elevated serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels. Transcriptional levels of apoptosis-related genes and toll-like receptor signaling pathway components were significantly up-regulated in liver under cold stress. Compared with control group, in terms of thermal stress, the growth performance and feed utilization of fish were declined to some extent compared with MT group. Moreover, high temperature significantly elevated hepatic productions of malondialdehyde and hydrogen peroxide, as well as increased activities of some antioxidant enzymes in liver. In addition, low and high temperature induce changes in the composition of gut microbiota. Overall, the results suggested that cold stress decelerated growth performance, induced hepatocyte apoptosis, and enhanced innate immunity in hybrid sturgeon to cope with additional stressors. Whereas, thermal stress resulted in hepatic oxidative stress in liver and the protective responses in the antioxidant enzymes in fish were activated. These results provided insights into the different physiological adaptation strategies in responsive to cold stress and thermal stress in this cold-water fish.
Collapse
Affiliation(s)
- Tianyu Liu
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Ling Li
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Yanchao Yang
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Jiarou Li
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Xutong Yang
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Lei Li
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Ziyao Zheng
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Boyu Yang
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Peiyu Zhang
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China; Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Shijiazhuang, 050024, China; Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, 050024, China.
| | - Haiyan Liu
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China; Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Shijiazhuang, 050024, China; Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, 050024, China.
| |
Collapse
|
29
|
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
|
30
|
Janiszewska J, Kostrzewska-Poczekaj M, Wierzbicka M, Brenner JC, Giefing M. HPV-driven oncogenesis-much more than the E6 and E7 oncoproteins. J Appl Genet 2025; 66:63-71. [PMID: 38907809 PMCID: PMC11761861 DOI: 10.1007/s13353-024-00883-y] [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: 11/28/2023] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/24/2024]
Abstract
High-risk human papillomaviruses are well-established drivers of several cancer types including cervical, head and neck, penile as well as anal cancers. While the E6 and E7 viral oncoproteins have proven to be critical for malignant transformation, evidence is also beginning to emerge suggesting that both host pathways and additional viral genes may also be pivotal for malignant transformation. Here, we focus on the role of host APOBEC genes, which have an important role in molecular editing including in the response to the viral DNA and their role in HPV-driven carcinogenesis. Further, we also discuss data developed suggesting the existence of HPV-derived miRNAs in HPV + tumors and their potential role in regulating the host transcriptome. Collectively, while recent advances in these two areas have added complexity to the working model of papillomavirus-induced oncogenesis, these discoveries have also shed a light onto new areas of research that will be required to fully understand the process.
Collapse
Affiliation(s)
- J Janiszewska
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
| | - M Kostrzewska-Poczekaj
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
| | - M Wierzbicka
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
- Research & Development Centre, Regional Specialist Hospital Wroclaw, Wroclaw, Poland
- Faculty of Medicine, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - J C Brenner
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - M Giefing
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland.
| |
Collapse
|
31
|
Lazure F, Drapela S, Liu X, Lockhart JH, Kashfi H, Sarigul N, Ilter D, Flores ER, Wang X, Smalley I, Jaeger A, Yu X, Gomes AP. Aging directs the differential evolution of KRAS-driven lung adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.20.633951. [PMID: 39896625 PMCID: PMC11785146 DOI: 10.1101/2025.01.20.633951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Lung adenocarcinoma (LUAD), the most common histological subtype of lung cancer(1, 2), is a disease of the elderly, with an average age of diagnosis of about 70 years of age(3). Older age is associated with an increased incidence of KRAS-driven LUAD(4), a particularly deadly type of LUAD characterized by treatment resistance and relapse. Despite this, our understanding of how old age shapes KRAS-driven LUAD evolution remains incomplete. While the age-related increase in cancer risk was previously ascribed to the accumulation of mutations over time, we are now beginning to consider the role of host biology as an independent factor influencing cancer. Here, we use single-cell RNA-Sequencing of KP (KrasG12D/+; Trp53flox/flox) LUAD transplanted into young and old mice to define how old age affects LUAD evolution and map the changes that old age imposes onto LUAD's microenvironment. Our data demonstrates that the aged lung environment steers LUAD evolution towards a primitive stem-like state that is associated with poor prognosis. We ascribe this differential evolution, at least in part, to a population of rare and highly secretory damage-associated alveolar differentiation intermediate (ADI) cells that accumulate in the aged tumor microenvironment (TME) and that dominate the niche signaling received by LUAD cells. Overall, our data puts aging center stage in coordinating LUAD evolution, highlighting the need to model LUAD in its most common context and creating a framework to tailor future cancer therapeutic strategies to the age of the patient to improve outcomes in the largest and most vulnerable LUAD patient population, the elderly.
Collapse
Affiliation(s)
- Felicia Lazure
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Stanislav Drapela
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Xiaoxian Liu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - John H Lockhart
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Hossein Kashfi
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Nadir Sarigul
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Didem Ilter
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Elsa R Flores
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Inna Smalley
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Alex Jaeger
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Ana P Gomes
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
32
|
Zhou X, Zhu X, Wang W, Wang J, Wen H, Zhao Y, Zhang J, Xu Q, Zhao Z, Ni T. Comprehensive Cellular Senescence Evaluation to Aid Targeted Therapies. RESEARCH (WASHINGTON, D.C.) 2025; 8:0576. [PMID: 39822281 PMCID: PMC11735710 DOI: 10.34133/research.0576] [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: 10/08/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/19/2025]
Abstract
Drug resistance to a single agent is common in cancer-targeted therapies, and rational drug combinations are a promising approach to overcome this challenge. Many Food and Drug Administration-approved drugs can induce cellular senescence, which possesses unique vulnerabilities and molecular signatures. However, there is limited analysis on the effect of the combination of cellular-senescence-inducing drugs and targeted therapy drugs. Here, we conducted a comprehensive evaluation of cellular senescence using 7 senescence-associated gene sets. We quantified the cellular senescence states of ~10,000 tumor samples from The Cancer Genome Atlas and examined their associations with targeted drug responses. Our analysis revealed that tumors with higher cellular senescence scores exhibited increased sensitivity to targeted drugs. As a proof of concept, we experimentally confirmed that etoposide-induced senescence sensitized lung cancer cells to 2 widely used targeted drugs, erlotinib and dasatinib. Furthermore, we identified multiple genes whose dependencies were associated with senescence status across ~1,000 cancer cell lines, suggesting that cellular senescence generates unique vulnerabilities for therapeutic exploitation. Our study provides a comprehensive overview of drug response related to cellular senescence and highlights the potential of combining senescence-inducing agents with targeted therapies to improve treatment outcomes in lung cancer, revealing novel applications of cellular senescence in targeted cancer therapies.
Collapse
Affiliation(s)
- Xiaolan Zhou
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences,
Fudan University, Shanghai 200438, China
| | - Xiaofeng Zhu
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences,
Fudan University, Shanghai 200438, China
| | - Weixu Wang
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences,
Fudan University, Shanghai 200438, China
- Institute of Computational Biology,
Helmholtz Center Munich, Munich, Germany
| | - Jing Wang
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences,
Fudan University, Shanghai 200438, China
| | - Haimei Wen
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences,
Fudan University, Shanghai 200438, China
| | - Yuqi Zhao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences,
Inner Mongolia University, Hohhot 010070, China
| | - Jiayu Zhang
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences,
Fudan University, Shanghai 200438, China
| | - Qiushi Xu
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences,
Fudan University, Shanghai 200438, China
| | - Zhaozhao Zhao
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences,
Fudan University, Shanghai 200438, China
- MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences,
Fudan University, Shanghai 200438, China
| | - Ting Ni
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences,
Fudan University, Shanghai 200438, China
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences,
Inner Mongolia University, Hohhot 010070, China
| |
Collapse
|
33
|
Wiechens E, Vigliotti F, Siniuk K, Schwarz R, Schwab K, Riege K, van Bömmel A, Görlich I, Bens M, Sahm A, Groth M, Sammons MA, Loewer A, Hoffmann S, Fischer M. Gene regulation by convergent promoters. Nat Genet 2025; 57:206-217. [PMID: 39779959 PMCID: PMC11735407 DOI: 10.1038/s41588-024-02025-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/04/2024] [Indexed: 01/11/2025]
Abstract
Convergent transcription, that is, the collision of sense and antisense transcription, is ubiquitous in mammalian genomes and believed to diminish RNA expression. Recently, antisense transcription downstream of promoters was found to be surprisingly prevalent. However, functional characteristics of affected promoters are poorly investigated. Here we show that convergent transcription marks an unexpected positively co-regulated promoter constellation. By assessing transcriptional dynamic systems, we identified co-regulated constituent promoters connected through a distinct chromatin structure. Within these cis-regulatory domains, transcription factors can regulate both constituting promoters by binding to only one of them. Convergent promoters comprise about a quarter of all active transcript start sites and initiate 5'-overlapping antisense RNAs-an RNA class believed previously to be rare. Visualization of nascent RNA molecules reveals convergent cotranscription at these loci. Together, our results demonstrate that co-regulated convergent promoters substantially expand the cis-regulatory repertoire, reveal limitations of the transcription interference model and call for adjusting the promoter concept.
Collapse
Affiliation(s)
- Elina Wiechens
- Hoffmann Lab, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Flavia Vigliotti
- Department of Biology, Systems Biology of the Stress Response, Technical University of Darmstadt, Darmstadt, Germany
| | - Kanstantsin Siniuk
- Hoffmann Lab, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Robert Schwarz
- Hoffmann Lab, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Katjana Schwab
- Hoffmann Lab, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Konstantin Riege
- Hoffmann Lab, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Alena van Bömmel
- Hoffmann Lab, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Ivonne Görlich
- Core Facility Next Generation Sequencing, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Martin Bens
- Core Facility Next Generation Sequencing, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Arne Sahm
- Hoffmann Lab, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
- Computational Phenomics Group, IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
- Computational Phenomics Group, Ruhr University Bochum, Bochum, Germany
| | - Marco Groth
- Core Facility Next Generation Sequencing, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Morgan A Sammons
- Department of Biological Sciences, The RNA Institute, The State University of New York at Albany, Albany, NY, USA
| | - Alexander Loewer
- Department of Biology, Systems Biology of the Stress Response, Technical University of Darmstadt, Darmstadt, Germany
| | - Steve Hoffmann
- Hoffmann Lab, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany.
| | - Martin Fischer
- Hoffmann Lab, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany.
| |
Collapse
|
34
|
Kim DE, Oh HJ, Kim HJ, Kim YB, Kim ST, Yim H. Synergistic two-step inhibition approach using a combination of trametinib and onvansertib in KRAS and TP53-mutated colorectal adenocarcinoma. Biomed Pharmacother 2025; 182:117796. [PMID: 39731938 DOI: 10.1016/j.biopha.2024.117796] [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: 12/17/2024] [Accepted: 12/21/2024] [Indexed: 12/30/2024] Open
Abstract
Colorectal malignancies associated with KRAS and TP53 mutations led us to investigate the effects of combination therapy targeting KRAS, MEK1, or PLK1 in colorectal cancer. MEK1 is downstream of RAS in the MAPK pathway, whereas PLK1 is a mitotic kinase of the cell cycle activated by MAPK and regulated by p53. Bioinformatics analysis revealed that patients with colorectal cancer had a high expression of MAP2K1 and PLK1. Furthermore, PLK1 and MEK1 activity in human colorectal adenocarcinoma (COAD) tissues was found to be highly upregulated compared to healthy tissues. To determine the sensitivity of KRAS or/and TP53-mutated cancer to KRAS, MEK1, or PLK1-targeted therapy, the inhibitors salirasib, trametinib, volasertib, and onvansertib were used in COAD cells with different KRAS and TP53 status. The results showed that combinations with trametinib and PLK1 inhibitors were more potent than combinations with salirasib. A combination of MEK1 and PLK1 inhibitors exhibited significant therapeutic effects on KRAS or/and TP53-mutated COAD cells. Notably, the combination of trametinib and onvansertib effectively suppressed tumor growth in a xenograft mouse model of KRAS and TP53-mutated COAD. This treatment induced G1 and G2/M arrest, respectively, and showed the strongest synergistic effect in KRAS and TP53-mutated SW48 cells expressing mutant KRASG13D and transduced with TP53 shRNA, ultimately leading to apoptotic cell death. These effects are attributed to two-step inhibition mechanism that blocks the MAPK signaling pathway and disrupts mitosis in KRAS and TP53-mutated COAD cells.
Collapse
Affiliation(s)
- Da-Eun Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Hyun-Ji Oh
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Hyun-Jin Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Yeo-Bin Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Seung-Tae Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Hyungshin Yim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea.
| |
Collapse
|
35
|
Levin-Ferreyra F, Kodali S, Cui Y, Pashos ARS, Pessina P, Brumbaugh J, Di Stefano B. Transposable element activity captures human pluripotent cell states. EMBO Rep 2025; 26:329-352. [PMID: 39668246 PMCID: PMC11772670 DOI: 10.1038/s44319-024-00343-y] [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: 08/04/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/14/2024] Open
Abstract
Human pluripotent stem cells (hPSCs) exist in multiple, transcriptionally distinct states and serve as powerful models for studying human development. Despite their significance, the molecular determinants and pathways governing these pluripotent states remain incompletely understood. Here, we demonstrate that transposable elements act as sensitive indicators of distinct pluripotent cell states. We engineered hPSCs with fluorescent reporters to capture the temporal expression dynamics of two state-specific transposable elements, LTR5_Hs, and MER51B. This dual reporter system enables real-time monitoring and isolation of stem cells transitioning from naïve to primed pluripotency and further towards differentiation, serving as a more accurate readout of pluripotency states compared to conventional systems. Unexpectedly, we identified a rare, metastable cell population within primed hPSCs, marked by transcripts related to preimplantation embryo development and which is associated with a DNA damage response. Moreover, our system establishes the chromatin factor NSD1 and the RNA-binding protein FUS as potent molecular safeguards of primed pluripotency. Our study introduces a novel system for investigating cellular potency and provides key insights into the regulation of embryonic development.
Collapse
Affiliation(s)
- Florencia Levin-Ferreyra
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Srikanth Kodali
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Yingzhi Cui
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Alison R S Pashos
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Patrizia Pessina
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Justin Brumbaugh
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Bruno Di Stefano
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA.
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
36
|
Ranganathan R, Li S, Sapozhnikov G, Wang S, Song YQ. Lower expression of BIN1's neuronal isoform in vulnerable excitatory neurons increases risk in Alzheimer's disease. J Alzheimers Dis Rep 2025; 9:25424823241296018. [PMID: 40034505 PMCID: PMC11864243 DOI: 10.1177/25424823241296018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/06/2024] [Indexed: 03/05/2025] Open
Abstract
Background Neurons in Alzheimer's disease (AD) experience elevated DNA damage, with DNA repair sites enriched at enhancer regions of genes essential for neuronal survival. Excitatory neurons in the cortical superficial layers expressing CUX2 and RORB (Cux2+/Rorb+), are selectively vulnerable in AD, but their relationship to single nucleotide polymorphisms (SNPs) in AD genome-wide association studies (GWAS) is unclear. Objective This study aimed to identify and characterize functional AD-GWAS SNPs using single-nucleus RNA sequencing data, focusing on selectively vulnerable neurons and DNA repair hotspots. Methods Filters were applied to identify candidate SNPs based on overlap with repair hotspots, RNA expression, transcription factor binding, AD association, and epigenetic significance. In vitro assays and analyses of large datasets from bulk RNA-seq (n = 1894), proteomics (n = 400), and single-nucleus RNA-seq (n = 424, 1.6 M cells) were conducted. Results BIN1 SNP, rs78710909, met multiple criteria - located in an AD-GWAS locus, repair hotspot, and promoter region. rs78710909C exhibits 1.52× higher AD risk and 5.4× differential transcription factor binding. In vitro, rs78710909C shows greater enhancer activity and weaker p53 but stronger E2F1 binding. BIN1's neuronal isoform is neuroprotective, but its AD expression is lower (p < 0.01). Moreover, only in AD and Cux2+/Rorb + neurons, rs78710909C is associated with a lower average BIN1 neuronal isoform ratio (p < 0.01). The genes upregulated in neurons with lower neuronal isoform ratio were associated with the hallmarks of AD pathology. Conclusions In a disease-relevant mechanism, the BIN1 SNP rs78710909C is associated with a lower ratio of BIN1's neuronal isoform which increases the vulnerability of specific excitatory neurons in AD patients.
Collapse
Affiliation(s)
- Rajesh Ranganathan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Siwen Li
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Georgy Sapozhnikov
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Shoutang Wang
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - You-Qiang Song
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
37
|
Janic A, Abad E, Amelio I. Decoding p53 tumor suppression: a crosstalk between genomic stability and epigenetic control? Cell Death Differ 2025; 32:1-8. [PMID: 38379088 PMCID: PMC11742645 DOI: 10.1038/s41418-024-01259-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/22/2024] Open
Abstract
Genomic instability, a hallmark of cancer, is a direct consequence of the inactivation of the tumor suppressor protein p53. Genetically modified mouse models and human tumor samples have revealed that p53 loss results in extensive chromosomal abnormalities, from copy number alterations to structural rearrangements. In this perspective article we explore the multifaceted relationship between p53, genomic stability, and epigenetic control, highlighting its significance in cancer biology. p53 emerges as a critical regulator of DNA repair mechanisms, influencing key components of repair pathways and directly participating in DNA repair processes. p53 role in genomic integrity however extends beyond its canonical functions. p53 influences also epigenetic landscape, where it modulates DNA methylation and histone modifications. This epigenetic control impacts the expression of genes involved in tumor suppression and oncogenesis. Notably, p53 ability to ensure cellular response to DNA demethylation contributes to the maintenance of genomic stability by preventing unscheduled transcription of repetitive non-coding genomic regions. This latter indicates a causative relationship between the control of epigenetic stability and the maintenance of genomic integrity in p53-mediated tumor suppression. Understanding these mechanisms offers promising avenues for innovative therapeutic strategies targeting epigenetic dysregulation in cancer and emphasizes the need for further research to unravel the complexities of this relationship. Ultimately, these insights hold the potential to transform cancer treatment and prevention strategies.
Collapse
Affiliation(s)
- Ana Janic
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
| | - Etna Abad
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ivano Amelio
- Chair for Systems Toxicology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
38
|
Zhang H, Zeng J, Zhang F, Liu J, Liang L. Role of B-Cell Lymphoma/Leukemia 11A in Normal and Malignant Hematopoiesis. BIOLOGY 2025; 14:26. [PMID: 39857257 PMCID: PMC11759832 DOI: 10.3390/biology14010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025]
Abstract
B-cell lymphoma/leukemia 11A (BCL11A) is a crucial transcriptional regulator, widely recognized for its role in controlling fetal hemoglobin and its potential as a gene therapy target for inherited hemoglobinopathies. Beyond this, recent studies have also highlighted its key role in the maturation and function of immune cells and erythrocytes, mediated through the regulation of various molecules during hematopoietic development. The dysregulation of BCL11A disrupts downstream molecular pathways, contributing to the development of several hematological malignancies, particularly leukemias. This review provides a comprehensive overview of the role of BCL11A in normal and malignant hematopoiesis, details the hematological disorders associated with its dysregulation and explores the current therapeutic strategies targeting this transcription factor.
Collapse
Affiliation(s)
- Haihang Zhang
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (H.Z.); (F.Z.)
| | - Junhao Zeng
- Xiangya School of Medicine, Central South University, Changsha 410013, China;
| | - Fangling Zhang
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (H.Z.); (F.Z.)
| | - Jing Liu
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (H.Z.); (F.Z.)
| | - Long Liang
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (H.Z.); (F.Z.)
| |
Collapse
|
39
|
Gong L, Xu D, Ni K, Li J, Mao W, Zhang B, Pu Z, Fang X, Yin Y, Ji L, Wang J, Hu Y, Meng J, Zhang R, Jiao J, Zou J. Smad1 Promotes Tumorigenicity and Chemoresistance of Glioblastoma by Sequestering p300 From p53. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2402258. [PMID: 39629919 PMCID: PMC11789598 DOI: 10.1002/advs.202402258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 06/26/2024] [Indexed: 01/30/2025]
Abstract
Acetylation is critically required for p53 activation, though it remains poorly understood how p53 acetylation is regulated in glioblastoma (GBM). This study reveals that p53 acetylation is a favorable prognostic marker for GBM, regardless of p53 status, and that Smad1, a key negative regulator of p53 acetylation, is involved in this process. Smad1 forms a complex with p53 and p300, inhibiting p300's interaction with p53 and leading to reduced p53 acetylation and increased Smad1 acetylation in GBM. This results in enhanced tumor growth and resistance to chemotherapy, particularly in tumors with missense mutant p53. Acetylation of K373 is found to be essential for Smad1's oncogenic function but does not confer chemoresistance in the absence of p53. Through molecular docking, it is discovered that Smad1 and p53 both interact with the acetyltransferase domain of p300, but at different amino acid sites. Disturbing the interface of Smad1 through amino acid mutations abolishes the Smad1-p300 complex and promotes p53 acetylation. Therefore, a small molecule is identified through virtual screening that specifically disrupts the Smad1-p300 interaction, offering a promising strategy for inhibiting GBM and increasing chemosensitivity by inhibiting Smad1 acetylation and restoring p53 acetylation.
Collapse
Affiliation(s)
- Lingli Gong
- Department of Laboratory MedicineThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's HospitalWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
- Wuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
| | - Daxing Xu
- Department of Laboratory MedicineThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's HospitalWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
- Wuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
| | - Kaixiang Ni
- Wuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
- Department of NeurosurgeryThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxiJiangsu214023China
| | - Jie Li
- Department of Laboratory MedicineThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's HospitalWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
- Wuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
| | - Wei Mao
- Department of Laboratory MedicineThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's HospitalWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
- Wuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
| | - Bo Zhang
- Department of Laboratory MedicineThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's HospitalWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
- Center of Clinical ResearchThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxiJiangsu214023China
| | - Zhening Pu
- Department of Laboratory MedicineThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's HospitalWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
- Center of Clinical ResearchThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxiJiangsu214023China
| | - Xiangming Fang
- Department of RadiologyThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxiJiangsu214023China
| | - Ying Yin
- Department of Laboratory MedicineThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's HospitalWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
- Wuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
| | - Li Ji
- Department of Laboratory MedicineThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's HospitalWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
- Wuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
| | - Jingjing Wang
- Department of Laboratory MedicineThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's HospitalWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
- Wuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
| | - Yaling Hu
- Department of Laboratory MedicineThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's HospitalWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
- Wuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
| | - Jiao Meng
- Department of Laboratory MedicineThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's HospitalWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
- Wuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
| | - Rui Zhang
- Department of NeurosurgeryThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxiJiangsu214023China
| | - Jiantong Jiao
- Wuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
- Department of NeurosurgeryThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxiJiangsu214023China
| | - Jian Zou
- Department of Laboratory MedicineThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's HospitalWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
- Wuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023China
| |
Collapse
|
40
|
Abdul Ghafoor N, Rasuli S, Tanriverdi Ö, Yildiz A. Investigating the P53-dependent anti-cancer effect of ibutamoren in human cancer cell lines. Basic Clin Pharmacol Toxicol 2025; 136:e14111. [PMID: 39668330 DOI: 10.1111/bcpt.14111] [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: 08/23/2024] [Revised: 10/29/2024] [Accepted: 11/19/2024] [Indexed: 12/14/2024]
Abstract
The MDM2-p53 pathway plays a pivotal role in regulating cell cycle and apoptosis, with its dysfunction contributing to approximately 50% of human malignancies. MDM2, an E3 ubiquitin ligase, targets the tumour suppressor p53 for degradation, thereby promoting uncontrolled cell growth in cancers. Inhibiting the MDM2-p53 interaction represents a promising therapeutic strategy for reactivating p53's tumour-suppressive functions. This study explored the potential of ibutamoren (IBU) as a novel inhibitor of MDM2. In silico analyses utilizing molecular modelling revealed that IBU has a low IC50 for MDM2 inhibition and favourably binds to the p53-binding pocket of MDM2. In vitro experiments demonstrated that IBU treatment reduced the viability of immortalized cancer cell lines with a functional MDM2-p53 pathway but not in cell lines where this pathway harboured damaging mutations. This trend was further supported by RT-qPCR analysis, which showed differential expression of two p53 target genes upon IBU treatment in cell lines with wild MDM2-p53 pathways but not in those harbouring damaging mutations. These findings provide preliminary evidence supporting IBU's anticancer activity, plausibly through the MDM2-p53 pathway, and suggest that further studies are warranted to explore its mechanism of action and potential development as a lead compound in oncology research.
Collapse
Affiliation(s)
- Naeem Abdul Ghafoor
- Department of Molecular Biology and Genetics, Graduate School of Natural and Applied Sciences, Mugla Sitki Kocman University, Mugla, Turkey
| | - Sabina Rasuli
- UFR Biosciences, Claude Bernard University Lyon 1, Villeurbanne, France
| | - Özgür Tanriverdi
- Department of Medical Oncology, Faculty of Medicine, Mugla Sitki Kocman University, Mugla, Turkey
| | - Ayşegül Yildiz
- Department of Molecular Biology and Genetics, Graduate School of Natural and Applied Sciences, Mugla Sitki Kocman University, Mugla, Turkey
- Department of Molecular Biology and Genetics, Faculty of Science, Mugla Sitki Kocman University, Mugla, Turkey
| |
Collapse
|
41
|
Magnani E, Macchi F, Randic T, Chen C, Madakashira B, Ranjan S, Eski SE, Singh SP, Sadler KC. Epigenetic Disordering Drives Stemness, Senescence Escape and Tumor Heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.29.629346. [PMID: 39763773 PMCID: PMC11703240 DOI: 10.1101/2024.12.29.629346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Tumor heterogeneity is the substrate for tumor evolution and the linchpin of treatment resistance. Cancer cell heterogeneity is largely attributed to distinct genetic changes within each cell population. However, the widespread epigenome repatterning that characterizes most cancers is also highly heterogenous within tumors and could generate cells with diverse identities and malignant features. We show that high levels of the epigenetic regulator and oncogene, UHRF1, in zebrafish hepatocytes rapidly induced methylome disordering, loss of heterochromatin, and DNA damage, resulting in cell cycle arrest, senescence, and acquisition of stemness. Reducing UHRF1 expression transitions these cells from senescent to proliferation-competent. The expansion of these damaged cells results in hepatocellular carcinomas (HCC) that have immature cancer cells intermingled with fibroblasts, immune and senescent cells expressing high UHRF1 levels, which serve as reservoirs for new cancer cells. This defines a distinct and heterogenous HCC subtype resulting from epigenetic changes, stemness and senescence escape.
Collapse
Affiliation(s)
- Elena Magnani
- Program in Biology, NYU Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| | - Filippo Macchi
- Program in Biology, NYU Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| | - Tijana Randic
- Program in Biology, NYU Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| | - Charlene Chen
- Program in Biology, NYU Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| | - Bhavani Madakashira
- Program in Biology, NYU Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| | - Shashi Ranjan
- Program in Biology, NYU Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| | - Sema Elif Eski
- Laboratory of Regeneration and Stress Biology, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM-Jacques E. Dumont), Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Sumeet P. Singh
- Laboratory of Regeneration and Stress Biology, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM-Jacques E. Dumont), Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Kirsten C. Sadler
- Program in Biology, NYU Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
- Center for Genomics and Systems Biology, NYU Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| |
Collapse
|
42
|
Mou K, Wang H, Zhu S, Luo J, Wang J, Peng L, Lei Y, Zhang Y, Huang S, Zhao H, Li G, Xiang L, Luo Y. Comprehensive analysis of the prognostic and immunological role of cavins in non-small cell lung cancer. BMC Cancer 2024; 24:1525. [PMID: 39695458 DOI: 10.1186/s12885-024-13280-9] [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: 12/19/2023] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
Caveolae, specialized and dynamic subdomains of the plasma membrane, have a crucial role in diverse cellular functions encompassing endocytosis, signal transduction, mechanosensation, lipid storage, and metabolism. Cavin family proteins are indispensable for caveolar formation and function. An increasing number of studies have found that cavins are involved in tumor growth, invasion, metastasis, and angiogenesis and may have dual roles in the regulation of cancer. However, the expression and prognostic value of cavins in non-small cell lung cancer (NSCLC) remain unexplored. In this study, the expression, survival data, immune infiltration, and functional enrichment of cavins in patients with NSCLC were investigated using multiple databases. Furthermore, different subtypes of cavin-binding proteins were identified through protein-protein interaction networks and k-means clustering. The results showed that the expression of Cavin-1-3 in NSCLC tissues was significantly lower than that in normal tissues, and that Cavin-2 is the major subtype of cavin that inhibits NSCLC progression. It regulates downstream signaling pathways, modulates the infiltration of immune cells and influences the prognosis of NSCLC. Related experiments also confirmed that Cavin-2 promotes the proliferation and metastasis of NSCLC cells. These findings suggest that cavins and their binding proteins may be novel biomarkers for NSCLC prognosis and immunotherapy, providing new treatment options for NSCLC.
Collapse
Affiliation(s)
- Kelin Mou
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Huan Wang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Siqi Zhu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jing Luo
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jianmei Wang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lin Peng
- Department of Bone and Joint, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yulin Lei
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yunke Zhang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shike Huang
- Department of Oncology, Hejiang County People's Hospital, Luzhou, China
| | - Huarong Zhao
- Department of Oncology, Hejiang County People's Hospital, Luzhou, China
| | - Gang Li
- Department of Oncology, Luzhou People's Hospital, Luzhou, China
| | - Li Xiang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
| | - Yuhao Luo
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
43
|
Li Q, Zhang Y, Luo S, Zhang Z, Oberg AL, Kozono DE, Lu H, Sarkaria JN, Ma L, Wang L. Identify Non-mutational p53 Functional Deficiency in Human Cancers. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae064. [PMID: 39325855 PMCID: PMC11702981 DOI: 10.1093/gpbjnl/qzae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 07/23/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024]
Abstract
An accurate assessment of p53's functional statuses is critical for cancer genomic medicine. However, there is a significant challenge in identifying tumors with non-mutational p53 inactivation which is not detectable through DNA sequencing. These undetected cases are often misclassified as p53-normal, leading to inaccurate prognosis and downstream association analyses. To address this issue, we built the support vector machine (SVM) models to systematically reassess p53's functional statuses in TP53 wild-type (TP53WT) tumors from multiple The Cancer Genome Atlas (TCGA) cohorts. Cross-validation demonstrated the good performance of the SVM models with a mean area under the receiver operating characteristic curve (AUROC) of 0.9822, precision of 0.9747, and recall of 0.9784. Our study revealed that a significant proportion (87%-99%) of TP53WT tumors actually had compromised p53 function. Additional analyses uncovered that these genetically intact but functionally impaired (termed as predictively reduced function of p53 or TP53WT-pRF) tumors exhibited genomic and pathophysiologic features akin to TP53-mutant tumors: heightened genomic instability and elevated levels of hypoxia. Clinically, patients with TP53WT-pRF tumors experienced significantly shortened overall survival or progression-free survival compared to those with predictively normal function of p53 (TP53WT-pN) tumors, and these patients also displayed increased sensitivity to platinum-based chemotherapy and radiation therapy.
Collapse
Affiliation(s)
- Qianpeng Li
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Zhang
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sicheng Luo
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhang Zhang
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ann L Oberg
- Division of Computational Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - David E Kozono
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, Boston, MA 02215, USA
| | - Hua Lu
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Lina Ma
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liguo Wang
- Division of Computational Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
- Bioinformatics and Computational Biology Graduate Program, University of Minnesota Rochester, Rochester, MN 55904, USA
| |
Collapse
|
44
|
Tatarata QZ, Wang Z, Konopleva M. BCL-2 inhibition in acute myeloid leukemia: resistance and combinations. Expert Rev Hematol 2024; 17:935-946. [PMID: 39552410 DOI: 10.1080/17474086.2024.2429604] [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: 06/19/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
INTRODUCTION The introduction of venetoclax has revolutionized the treatment landscape of acute myeloid leukemia, offering new therapeutic opportunities. However, the clinical response to venetoclax varies significantly between patients, with many experiencing limited duration of response. AREAS COVERED Identified resistance mechanisms include both intrinsic and acquired resistance to VEN. The former is associated with cell lineage and differentiation state. The latter includes dependency on alternative BCL-2 family anti-apoptotic protein(s) mediated by genetic, epigenetic, or post-translational mechanisms, mitochondrial and metabolic involvement, as well as microenvironment. Understanding these mechanisms is crucial for optimizing venetoclax-based therapies and enhancing treatment outcomes for patients with acute myeloid leukemia. This review aims to elucidate the primary mechanisms underlying resistance to venetoclax and explore current therapeutic strategies to overcome this challenge. EXPERT OPINION In patients with venetoclax resistance, alternative options include targeted combination therapies tailored to individual cases based on cytogenetics and prior treatments. Many of these therapies require further clinical investigation to validate their safety and efficacy.
Collapse
Affiliation(s)
- Qi Zhang Tatarata
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
- The Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Zhe Wang
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
- Department of Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
45
|
Perez AA, Goronzy IN, Blanco MR, Yeh BT, Guo JK, Lopes CS, Ettlin O, Burr A, Guttman M. ChIP-DIP maps binding of hundreds of proteins to DNA simultaneously and identifies diverse gene regulatory elements. Nat Genet 2024; 56:2827-2841. [PMID: 39587360 DOI: 10.1038/s41588-024-02000-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 10/21/2024] [Indexed: 11/27/2024]
Abstract
Gene expression is controlled by dynamic localization of thousands of regulatory proteins to precise genomic regions. Understanding this cell type-specific process has been a longstanding goal yet remains challenging because DNA-protein mapping methods generally study one protein at a time. Here, to address this, we developed chromatin immunoprecipitation done in parallel (ChIP-DIP) to generate genome-wide maps of hundreds of diverse regulatory proteins in a single experiment. ChIP-DIP produces highly accurate maps within large pools (>160 proteins) for all classes of DNA-associated proteins, including modified histones, chromatin regulators and transcription factors and across multiple conditions simultaneously. First, we used ChIP-DIP to measure temporal chromatin dynamics in primary dendritic cells following LPS stimulation. Next, we explored quantitative combinations of histone modifications that define distinct classes of regulatory elements and characterized their functional activity in human and mouse cell lines. Overall, ChIP-DIP generates context-specific protein localization maps at consortium scale within any molecular biology laboratory and experimental system.
Collapse
Affiliation(s)
- Andrew A Perez
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Isabel N Goronzy
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Mario R Blanco
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Benjamin T Yeh
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jimmy K Guo
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Carolina S Lopes
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Olivia Ettlin
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Alex Burr
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Mitchell Guttman
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
46
|
Kukreja K, Jia BZ, McGeary SE, Patel N, Megason SG, Klein AM. Cell state transitions are decoupled from cell division during early embryo development. Nat Cell Biol 2024; 26:2035-2045. [PMID: 39516639 DOI: 10.1038/s41556-024-01546-0] [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: 10/10/2023] [Accepted: 09/23/2024] [Indexed: 11/16/2024]
Abstract
As tissues develop, cells divide and differentiate concurrently. Conflicting evidence shows that cell division is either dispensable or required for formation of cell types. Here, to determine the role of cell division in differentiation, we arrested the cell cycle in zebrafish embryos using two independent approaches and profiled them at single-cell resolution. We show that cell division is dispensable for differentiation of all embryonic tissues from early gastrulation to the end of segmentation. However, arresting cell division does slow down differentiation in some cell types, and it induces global stress responses. While differentiation is robust to blocking cell division, the proportions of cells across cell states are not, but show evidence of partial compensation. This work clarifies our understanding of the role of cell division in development and showcases the utility of combining embryo-wide perturbations with single-cell RNA sequencing to uncover the role of common biological processes across multiple tissues.
Collapse
Affiliation(s)
- Kalki Kukreja
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Bill Z Jia
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Sean E McGeary
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Nikit Patel
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sean G Megason
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Allon M Klein
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
47
|
Tomkova K, Roman M, Adebayo AS, Sheikh S, Yusoff S, Gulston M, Joel-David L, Lai FY, Murgia A, Eagle-Hemming B, Aujla H, Chad T, Richardson GD, Griffin JL, Murphy GJ, Woźniak MJ. Multimorbidity is associated with myocardial DNA damage, nucleolar stress, dysregulated energy metabolism, and senescence in cardiovascular disease. NPJ AGING 2024; 10:58. [PMID: 39604391 PMCID: PMC11603063 DOI: 10.1038/s41514-024-00183-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024]
Abstract
This study investigates why individuals with multimorbidity-two or more chronic conditions-are more prone to adverse outcomes after surgery. In our cohort, ninety-eight of 144 participants had multimorbidity. The myocardial transcriptome and metabolites involved in energy production were measured in 53 and 57 sequential participants, respectively. Untargeted analysis of the metabolome in blood and myocardium was performed in 30 sequential participants. Mitochondrial respiration in circulating mononuclear cells was measured in 70 participants. Results highlighted four main biological processes associated with multimorbidity: DNA damage with epigenetic changes, mitochondrial energy disruption, cellular aging (senescence) and innate immune response. Histone 2B, its ubiquitination enzymes and AKT3 were upregulated in the multimorbid group. Plasma senescence-associated proteins (IL-1β, GM-CSF) increased with more comorbidities. DNA damage and nucleolar instability were specifically apparent in multimorbid myocardium. We conclude that multimorbidity in cardiovascular patients accelerates biological aging, making them more vulnerable to metabolic stress.
Collapse
Affiliation(s)
- Kristina Tomkova
- Department of Cardiovascular Sciences and NIHR Cardiovascular Biomedical Research Unit, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Marius Roman
- Department of Cardiovascular Sciences and NIHR Cardiovascular Biomedical Research Unit, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Adewale S Adebayo
- Department of Cardiovascular Sciences and NIHR Cardiovascular Biomedical Research Unit, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Sophia Sheikh
- Department of Cardiovascular Sciences and NIHR Cardiovascular Biomedical Research Unit, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Syabira Yusoff
- Department of Cardiovascular Sciences and NIHR Cardiovascular Biomedical Research Unit, University of Leicester, Glenfield Hospital, Leicester, UK
- Cardiovascular Sciences, King's College London, London, UK
| | - Melanie Gulston
- Department of Biochemistry and Cambridge Systems Biology Centre, The Sanger Building, 80 Tennis Court Road, Cambridge, UK
| | - Lathishia Joel-David
- Department of Cardiovascular Sciences and NIHR Cardiovascular Biomedical Research Unit, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Florence Y Lai
- Department of Cardiovascular Sciences and NIHR Cardiovascular Biomedical Research Unit, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Antonio Murgia
- Department of Biochemistry and Cambridge Systems Biology Centre, The Sanger Building, 80 Tennis Court Road, Cambridge, UK
| | - Bryony Eagle-Hemming
- Department of Cardiovascular Sciences and NIHR Cardiovascular Biomedical Research Unit, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Hardeep Aujla
- Department of Cardiovascular Sciences and NIHR Cardiovascular Biomedical Research Unit, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Tom Chad
- Department of Cardiovascular Sciences and NIHR Cardiovascular Biomedical Research Unit, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Gavin D Richardson
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Julian L Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, The Sanger Building, 80 Tennis Court Road, Cambridge, UK
- University of Aberdeen, King's College, Aberdeen, UK
| | - Gavin J Murphy
- Department of Cardiovascular Sciences and NIHR Cardiovascular Biomedical Research Unit, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Marcin J Woźniak
- Department of Cardiovascular Sciences and NIHR Cardiovascular Biomedical Research Unit, University of Leicester, Glenfield Hospital, Leicester, UK.
| |
Collapse
|
48
|
Geraghty S, Boyer JA, Fazel-Zarandi M, Arzouni N, Ryseck RP, McBride MJ, Parsons LR, Rabinowitz JD, Singh M. Integrative Computational Framework, Dyscovr, Links Mutated Driver Genes to Expression Dysregulation Across 19 Cancer Types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624509. [PMID: 39605479 PMCID: PMC11601522 DOI: 10.1101/2024.11.20.624509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Though somatic mutations play a critical role in driving cancer initiation and progression, the systems-level functional impacts of these mutations-particularly, how they alter expression across the genome and give rise to cancer hallmarks-are not yet well-understood, even for well-studied cancer driver genes. To address this, we designed an integrative machine learning model, Dyscovr, that leverages mutation, gene expression, copy number alteration (CNA), methylation, and clinical data to uncover putative relationships between nonsynonymous mutations in key cancer driver genes and transcriptional changes across the genome. We applied Dyscovr pan-cancer and within 19 individual cancer types, finding both broadly relevant and cancer type-specific links between driver genes and putative targets, including a subset we further identify as exhibiting negative genetic relationships. Our work newly implicates-and validates in cell lines-KBTBD2 and mutant PIK3CA as putative synthetic lethals in breast cancer, suggesting a novel combinatorial treatment approach.
Collapse
Affiliation(s)
- Sara Geraghty
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544
| | - Jacob A. Boyer
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544
- Ludwig Cancer Institute, Princeton Branch, Princeton University, Princeton, NJ 08554
| | - Mahya Fazel-Zarandi
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544
| | - Nibal Arzouni
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544
| | - Rolf-Peter Ryseck
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544
| | - Matthew J. McBride
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854
| | - Lance R. Parsons
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544
| | - Joshua D. Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544
- Ludwig Cancer Institute, Princeton Branch, Princeton University, Princeton, NJ 08554
- Department of Chemistry, Princeton University, Princeton, NJ 08544
| | - Mona Singh
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544
- Department of Computer Science, Princeton University, Princeton, NJ 08544
- Lead Contact
| |
Collapse
|
49
|
Klein SM, Bozko M, Toennießen A, Rangno D, Bozko P. High p53 Protein Level Is a Negative Prognostic Marker for Pancreatic Adenocarcinoma. Int J Mol Sci 2024; 25:12307. [PMID: 39596373 PMCID: PMC11594790 DOI: 10.3390/ijms252212307] [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: 09/25/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Pancreatic adenocarcinoma is one of the most aggressive types of cancer. Among different mechanisms generally believed to be important for the development of cancer, aberrant regulation of the p53 protein is a well-known and common feature for many cancer entities. Our work aims to analyze the impact of p53 deregulation and proteins encoded by p53 target genes on the survival of patients suffering from pancreatic adenocarcinoma. We, therefore, focused on the analysis of the selected collective for the TP53 mutation status, the p53 protein level, their correlation, and possible impacts on the prognosis/survival. We compared and analyzed a set of 123 patients. We have extracted information regarding the TP53 mutation status, p53 protein levels, the level of proteins encoded by prominent p53 target genes, and information on the overall survival. Survival analyses were displayed by Kaplan-Meier plots, using the log-rank test, in order to check for statistical significance. Protein levels were compared using the Mann-Whitney Test. We did not find any statistically significant correlation between the TP53 mutation status and the survival of the patients. Moreover, we have not found any significant correlation between the protein amount of prominent p53 target genes and the patients' survival. However, we see a significant correlation between the p53 protein level in cancer samples and the overall survival of pancreatic adenocarcinoma patients: patients having tumors with a p53 protein level within the upper quartile of all measured cases show a significantly reduced survival compared to the rest of the patients. Thus, in pancreatic adenocarcinoma, the p53 protein level is a relevant marker for prognosis, and cancers having a high p53 protein amount show a shortened patients' survival. In contrast, for this cancer entity, the TP53 mutation status or the protein amount of prominent p53 target genes on their own seems not to have a significant impact on survival.
Collapse
Affiliation(s)
- Sebastian M. Klein
- Department of Internal Medicine I, Universitätsklinikum Tübingen, Universität Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
- M3 Research Institute, University Tübingen, 72076 Tübingen, Germany
| | - Maria Bozko
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, 02-106 Warsaw, Poland
| | | | - Dennis Rangno
- Department of Internal Medicine I, Universitätsklinikum Tübingen, Universität Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Przemyslaw Bozko
- Department of Internal Medicine I, Universitätsklinikum Tübingen, Universität Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
- M3 Research Institute, University Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
50
|
Peng Y, Bai J, Li W, Su Z, Cheng X. Advancements in p53-Based Anti-Tumor Gene Therapy Research. Molecules 2024; 29:5315. [PMID: 39598704 PMCID: PMC11596491 DOI: 10.3390/molecules29225315] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
The p53 gene is one of the genes most closely associated with human tumors and has become a popular target for tumor drug design. Currently, p53-based gene therapy techniques have been developed, but these therapies face challenges such as immaturity, high safety hazards, limited efficacy, and low patient acceptance. However, researchers are no less enthusiastic about the treatment because of its theoretical potential to treat cancer. In this paper, the advances in p53-based gene therapy and related nucleic acid delivery technologies were reviewed and prospected in order to support further development in this field.
Collapse
Affiliation(s)
- Yuanwan Peng
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| | - Jinping Bai
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| | - Wang Li
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| | - Zhengding Su
- School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| | - Xiyao Cheng
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| |
Collapse
|