1
|
Tseng YH, Tran TTM, Tsai Chang J, Huang YT, Nguyen AT, Chang IYF, Chen YT, Hsieh HW, Juang YL, Chang PMH, Huang TY, Chang YC, Chen YM, Liu H, Huang CYF. Utilizing TP53 hotspot mutations as effective predictors of gemcitabine treatment outcome in non-small-cell lung cancer. Cell Death Discov 2025; 11:26. [PMID: 39870629 PMCID: PMC11772833 DOI: 10.1038/s41420-025-02300-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 12/11/2024] [Accepted: 01/14/2025] [Indexed: 01/29/2025] Open
Abstract
TP53 mutations are recognized to correlate with a worse prognosis in individuals with non-small cell lung cancer (NSCLC). There exists an immediate necessity to pinpoint selective treatment for patients carrying TP53 mutations. Potential drugs were identified by comparing drug sensitivity differences, represented by the half-maximal inhibitory concentration (IC50), between TP53 mutant and wild-type NSCLC cell lines using database analysis. In addition, clinical data from NSCLC patients were collected to evaluate both their TP53 status and their response to gemcitabine, thereby facilitating further validation. Subsequently, NSCLC cell lines with different TP53 status (A549 and H1299) were subjected to gemcitabine treatment to investigate the association between TP53 mutations and gemcitabine response. According to the dataset, NSCLC cell lines carrying TP53 mutations displayed heightened sensitivity to gemcitabine. From a clinical standpoint, patients exhibiting TP53 hotspot mutations demonstrated prolonged overall survival upon gemcitabine treatment. In vitro, overexpressing various hotspot TP53 mutations significantly sensitized H1299 cells to gemcitabine. Moreover, the knockdown of TP53 in A549 cells notably augmented sensitivity to gemcitabine treatment, as evidenced by cell viability and reproductive cell death assays. Conversely, the overexpression of wild-type TP53 in H1299 cells led to an increased resistance against gemcitabine. Gemcitabine is a treatment option for patients with non-small cell lung cancer (NSCLC) who carry TP53 hotspot mutations. This potential effectiveness might arise from its ability to disrupt DNA damage repair processes, leading to G2/M phase cell cycle arrest or an augmentation of mitotic abnormalities, eventually cause cell death. As a result, when planning treatment strategies for NSCLC patients possessing TP53 hotspot mutations, gemcitabine should be considered to incorporate into the indication.
Collapse
Affiliation(s)
- Yen-Han Tseng
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Program in Molecular Medicine, School of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Trieu Thi My Tran
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jinghua Tsai Chang
- Institute of Medicine, Chung Shan Medical University, Taichung City, Taiwan
| | - Yu-Tang Huang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Biomedical Industry Ph.D. Program, National Yang Ming Chiao Tung University, Hsinchu City, Taiwan
| | - Anh Thuc Nguyen
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Taiwan National Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Ian Yi-Feng Chang
- Taiwan National Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Tung Chen
- Taiwan National Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Hao-Wen Hsieh
- Department of Neurosurgery, Lin-Kou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yue-Li Juang
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Peter Mu-Hsin Chang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzu-Yi Huang
- Program in Molecular Medicine, School of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ying-Chih Chang
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Yuh-Min Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Hsuan Liu
- Biomedical Industry Ph.D. Program, National Yang Ming Chiao Tung University, Hsinchu City, Taiwan.
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Chong Hin Loon Memorial Cancer and Biotherapy Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
2
|
Doghish AS, Mageed SSA, Zaki MB, Abd-Elmawla MA, Sayed GA, Hatawsh A, Aborehab NM, Moussa R, Mohammed OA, Abdel-Reheim MA, Elimam H. Role of long non-coding RNAs and natural products in prostate cancer: insights into key signaling pathways. Funct Integr Genomics 2025; 25:16. [PMID: 39821470 DOI: 10.1007/s10142-025-01526-z] [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: 11/05/2024] [Revised: 12/18/2024] [Accepted: 01/01/2025] [Indexed: 01/19/2025]
Abstract
Prostate cancer (PC) ranks among the most prevalent cancers in males. Recent studies have highlighted intricate connections between long non-coding RNAs (lncRNAs), natural products, and cellular signaling in PC development. LncRNAs, which are RNA transcripts without protein-coding function, influence cell growth, programmed cell death, metastasis, and resistance to treatments through pathways like PI3K/AKT, WNT/β-catenin, and androgen receptor signaling. Certain lncRNAs, including HOTAIR and PCA3, are associated with PC progression, with potential as diagnostic markers. Natural compounds, such as curcumin and resveratrol, demonstrate anticancer effects by targeting these pathways, reducing tumor growth, and modulating lncRNA expression. For instance, curcumin suppresses HOTAIR levels, hindering PC cell proliferation and invasion. The interaction between lncRNAs and natural compounds may open new avenues for therapy, as these substances can simultaneously impact multiple signaling pathways. These complex interactions offer promising directions for developing innovative PC treatments, enhancing diagnostics, and identifying new biomarkers for improved prevention and targeted therapy. This review aims to map the multifaceted relationship among natural products, lncRNAs, and signaling pathways in PC pathogenesis, focusing on key pathways such as AR, PI3K/AKT/mTOR, WNT/β-catenin, and MAPK, which are crucial in PC progression and therapy resistance. Regulation of these pathways by natural products and lncRNAs could lead to new insights into biomarker identification, preventive measures, and targeted PC therapies.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt.
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt.
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, CairoE, 11829, Egypt
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ghadir A Sayed
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Cairo, 11829, Egypt
| | - Abdulrahman Hatawsh
- Biotechnology School, Nile University, 26th of July Corridor, Sheikh Zayed City, Giza, 12588, Egypt
| | - Nora M Aborehab
- Department of Biochemistry, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Rewan Moussa
- School Faculty of Medicine, Helwan University, Cairo, 11795, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia
| | | | - Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| |
Collapse
|
3
|
Luo C, Min X, Zhang D. New insights into the mechanisms of the immune microenvironment and immunotherapy in osteosarcoma. Front Immunol 2025; 15:1539696. [PMID: 39896817 PMCID: PMC11782189 DOI: 10.3389/fimmu.2024.1539696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025] Open
Abstract
Osteosarcoma, a malignant bone tumor primarily affecting adolescents, is highly invasive with a poor prognosis. While surgery and chemotherapy have improved survival for localized cases, pulmonary metastasis significantly reduces survival to approximately 20%, highlighting the need for novel treatments. Immunotherapy, which leverages the immune system to target osteosarcoma cells, shows promise. This review summarizes the biological characteristics of osteosarcoma, mechanisms of pulmonary metastasis, and the tumor immune microenvironment (TME). It involves recent immunotherapy advances, including monoclonal antibodies, tumor vaccines, immune cell therapies, checkpoint inhibitors, and oncolytic viruses, and discusses combining these with standard treatments.
Collapse
Affiliation(s)
- Cong Luo
- Department of Orthopedic Trauma, Zhuji People’s Hospital of Zhejiang Province, Zhuji, Zhejiang, China
| | - Xingxing Min
- Department of Orthopedic Trauma, Zhuji People’s Hospital of Zhejiang Province, Zhuji, Zhejiang, China
| | - Danying Zhang
- Department of Emergency and Critical Care, Shanghai Changzheng Hospital, Shanghai, China
| |
Collapse
|
4
|
Meng F, Qi T, Liu X, Wang Y, Yu J, Lu Z, Cai X, Li A, Jung D, Duan J. Enhanced pharmacological activities of AKR1C3-activated prodrug AST-3424 in cancer cells with defective DNA repair. Int J Cancer 2025; 156:417-430. [PMID: 39243400 DOI: 10.1002/ijc.35170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/28/2024] [Accepted: 08/07/2024] [Indexed: 09/09/2024]
Abstract
AST-3424 is a novel and highly tumor-selective prodrug. AST-3424 is activated by AKR1C3 to release a toxic bis-alkylating moiety, AST 2660. In this study, we have investigated the essential role of DNA repair in AST-3424 mediated pharmacological activities in vitro and in vivo. We show here that AST-3424 is effective as a single therapeutic agent against cancer cells to induce cytotoxicity, DNA damage, apoptosis and cell cycle arrest at G2 phase in a dose- and AKR1C3-dependent manner in both p53-proficient H460 (RRID:CVCL_0459) and p53-deficient HT-29 cells (RRID:CVCL_0320). The combination of abrogators of G2 checkpoint with AST-3424 was only synergistic in HT-29 but not in H460 cells. The enhanced activity of AST-3424 in HT-29 cells was due to impaired DNA repair ability via the attenuation of cell cycle G2 arrest and reduced RAD51 expression. Furthermore, we utilized a BRCA2 deficient cell line and two PDX models with BRCA deleterious mutations to study the increased activity of AST-3424. The results showed that AST-3424 exhibited enhanced in vitro cytotoxicity and superior and durable in vivo anti-tumor effects in cells deficient of DNA repair protein BRCA2. In summary, we report here that when DNA repair capacity is reduced, the in vitro and in vivo activity of AST-3424 can be further enhanced, thus providing supporting evidence for the further evaluation of AST-3424 in the clinic.
Collapse
Affiliation(s)
- Fanying Meng
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Tianyang Qi
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Xing Liu
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Yizhi Wang
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Jibing Yu
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Zhaoqiang Lu
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Xiaohong Cai
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Anrong Li
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Don Jung
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Jianxin Duan
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| |
Collapse
|
5
|
Liu J, Zhang B, Huang B, Zhang K, Guo F, Wang Z, Shang D. A stumbling block in pancreatic cancer treatment: drug resistance signaling networks. Front Cell Dev Biol 2025; 12:1462808. [PMID: 39872846 PMCID: PMC11770040 DOI: 10.3389/fcell.2024.1462808] [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: 07/10/2024] [Accepted: 12/30/2024] [Indexed: 01/30/2025] Open
Abstract
The primary node molecules in the cell signaling network in cancer tissues are maladjusted and mutated in comparison to normal tissues, which promotes the occurrence and progression of cancer. Pancreatic cancer (PC) is a highly fatal cancer with increasing incidence and low five-year survival rates. Currently, there are several therapies that target cell signaling networks in PC. However, PC is a "cold tumor" with a unique immunosuppressive tumor microenvironment (poor effector T cell infiltration, low antigen specificity), and targeting a single gene or pathway is basically ineffective in clinical practice. Targeted matrix therapy, targeted metabolic therapy, targeted mutant gene therapy, immunosuppressive therapy, cancer vaccines, and other emerging therapies have shown great therapeutic potential, but results have been disappointing. Therefore, we summarize the identified and potential drug-resistant cell signaling networks aimed at overcoming barriers to existing PC therapies.
Collapse
Affiliation(s)
- Jinming Liu
- Department of General Surgery, Pancreas and Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Biao Zhang
- Department of General Surgery, Pancreas and Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bingqian Huang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Department of Clinical Pharmacy, Affiliated Hangzhou First People’s Hospital, Westlake University, Hangzhou, China
| | - Kexin Zhang
- Central Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fujia Guo
- Central Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhizhou Wang
- Department of General Surgery, Pancreas and Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- Department of General Surgery, Pancreas and Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| |
Collapse
|
6
|
Spigarelli R, Spisni E, Magalhães M, Cabral C, Gonçalves AC, Saracino IM, Botti G, Dalpiaz A, Beggiato S, Valerii MC. Clove Essential Oil as a Source of Antitumoral Compounds Capable of Crossing the Blood-Brain Barrier: A Focus on the Effects of β-Caryophyllene and Eugenol in a Glioblastoma Cell Line. Int J Mol Sci 2024; 26:238. [PMID: 39796096 PMCID: PMC11720353 DOI: 10.3390/ijms26010238] [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: 12/08/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
This study aimed to investigate β-Caryophyllene (BCA) pharmacokinetics as well as the potential antitumor activity and mechanism of action of BCA and eugenol (EU), alone or in combination, in U87 glioblastoma (GB) cells. The BCA pharmacokinetic was studied by evaluating its concentration profiles in rat blood and cerebrospinal fluid after oral and intravenous administration. EU and BCA antitumor mechanisms were assessed by comparing their effects in U87 GB cells and non-tumoral HMC3 cells. Cell death, cell cycle regulation and mitochondrial membrane potential (MMP) were evaluated using flow cytometry. mRNA levels of target genes were evaluated by qPCR. Secreted cytokines were measured by Luminex®. BCA, as well as EU, permeates the brain. EU and BCA affected the viability and proliferation of U87 cells (up to 50%, p < 0.001) but not HMC3 cells and showed a synergistic effect. BCA and EU induced G0/G1 cell cycle arrest, increasing apoptosis/necrosis. EU and BCA induced the downregulation of mRNAs encoding for key proteins involved in GB angiogenesis (VEGFA decreased op to 60%, p < 0.01), proliferation and progression, and showed anti-inflammatory activity (IL-4 significantly decreased, p < 0.001). EU and BCA demonstrated strong and multitarget antitumor activity in U87 cells. Our results provide a strong rationale for the further evaluation of EU and BCA as possible therapeutic molecules in GB management.
Collapse
Affiliation(s)
- Renato Spigarelli
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Via Selmi 3, 40126 Bologna, Italy; (R.S.); (M.C.V.)
| | - Enzo Spisni
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Via Selmi 3, 40126 Bologna, Italy; (R.S.); (M.C.V.)
| | - Mariana Magalhães
- Institute for Interdisciplinary Research, Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, 3030-789 Coimbra, Portugal;
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Célia Cabral
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Centre for Functional Ecology, Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology, University Clinic of Hematology and Applied Molecular Biology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- iCBR, Group of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ilaria Maria Saracino
- Microbiology Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy;
| | - Giada Botti
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Fossato di Mortara 19, 44121 Ferrara, Italy; (G.B.); (A.D.)
| | - Alessandro Dalpiaz
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Fossato di Mortara 19, 44121 Ferrara, Italy; (G.B.); (A.D.)
| | - Sarah Beggiato
- Department of Life Sciences and Biotechnology, University of Ferrara and LTTA Center, Via Fossato di Mortara 19, 44121 Ferrara, Italy;
| | - Maria Chiara Valerii
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Via Selmi 3, 40126 Bologna, Italy; (R.S.); (M.C.V.)
| |
Collapse
|
7
|
Zhang B, Kapur P, Koduru PR, Jia L. Retroperitoneal Sarcomatoid Yolk Sac Tumor in a Chemotherapy-Naive Patient With Testicular Postpubertal Type Teratoma: A Rare Case Report With Emphasis on Molecular Features. Int J Surg Pathol 2024; 32:1537-1543. [PMID: 38377960 DOI: 10.1177/10668969241231973] [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] [Indexed: 02/22/2024]
Abstract
Sarcomatoid yolk sac tumor is a very rare histologic type of testicular germ cell tumor and is mainly reported in testicular germ cell tumor patients who receive chemotherapy. Herein, we report an extremely rare concurrent retroperitoneal sarcomatoid yolk sac tumor in a man with a testicular postpuberal teratoma before he received chemotherapy. A 37-year-old man initially presented with a persistent abdominal pain. Subsequent imaging studies revealed a 9.6-cm retroperitoneal mass, and 2 testicular masses (3.1 cm and 0.9 cm in greatest dimension, respectively). His serum tumor markers were within normal ranges. His radical orchiectomy demonstrated a postpubertal type teratoma with an adjacent scarring nodule. Later, his retroperitoneal tumor showed spindle tumor cells embedded in predominantly myxoid and focally fibrous stroma with diffuse and strong immunoreactivity for keratin AE1/AE3, SALL4 and glypican 3. No tumor necrosis or brisk mitotic figures were observed. A diagnosis of sarcomatoid yolk sac tumor was rendered. Fluorescence in situ hybridization analysis of his retroperitoneal sarcomatoid yolk sac tumor revealed polysomy 12 and MYC amplification, whereas no evidence of isochromosome 12p [i(12p)], and DNA sequencing showed 6 mutations per megabase (muts/Mb), and the somatic alterations included ARAF amplification and ATR I774Yfs*5. Considering its rarity, sarcomatoid yolk sac tumor may pose diagnostic challenges. Therefore, relevant clinicoradiologic information and ancillary work up, including immunohistochemistry and molecular studies, may be helpful for the accurate classification. Our tumor further raises awareness of this rare event, expands the spectrum of its clinical presentation, and explores the molecular features.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Prasad R Koduru
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Liwei Jia
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
8
|
Cordova RA, Sommers NR, Law AS, Klunk AJ, Brady KE, Goodrich DW, Anthony TG, Brault JJ, Pili R, Wek RC, Staschke KA. Coordination between the eIF2 kinase GCN2 and p53 signaling supports purine metabolism and the progression of prostate cancer. Sci Signal 2024; 17:eadp1375. [PMID: 39591412 DOI: 10.1126/scisignal.adp1375] [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: 03/29/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
Cancers invoke various pathways to mitigate external and internal stresses to continue their growth and progression. We previously reported that the eIF2 kinase GCN2 and the integrated stress response are constitutively active in prostate cancer (PCa) and are required to maintain amino acid homeostasis needed to fuel tumor growth. However, although loss of GCN2 function reduces intracellular amino acid availability and PCa growth, there is no appreciable cell death. Here, we discovered that the loss of GCN2 in PCa induces prosenescent p53 signaling. This p53 activation occurred through GCN2 inhibition-dependent reductions in purine nucleotides that impaired ribosome biogenesis and, consequently, induced the impaired ribosome biogenesis checkpoint. p53 signaling induced cell cycle arrest and senescence that promoted the survival of GCN2-deficient PCa cells. Depletion of GCN2 combined with loss of p53 or pharmacological inhibition of de novo purine biosynthesis reduced proliferation and enhanced cell death in PCa cell lines, organoids, and xenograft models. Our findings highlight the coordinated interplay between GCN2 and p53 regulation during nutrient stress and provide insight into how they could be targeted in developing new therapeutic strategies for PCa.
Collapse
Affiliation(s)
- Ricardo A Cordova
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Noah R Sommers
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Andrew S Law
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, USA
| | - Angela J Klunk
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Katherine E Brady
- Department of Biology, Indiana University School of Science, Indianapolis, IN 46202, USA
| | - David W Goodrich
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Tracy G Anthony
- Department of Nutritional Sciences and the New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ 08901, USA
| | - Jeffrey J Brault
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, USA
| | - Roberto Pili
- Jacobs School of Medicine and Biomedical Sciences, Division of Hematology and Oncology, University at Buffalo, Buffalo, NY 14203, USA
| | - Ronald C Wek
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Kirk A Staschke
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| |
Collapse
|
9
|
Nissenbaum J, Segal E, Philip H, Cashman R, Golan-Lev T, Reubinoff BE, Turjeman A, Yanuka O, Lezmi E, Kopper O, Benvenisty N. Predicting tumour resistance to paclitaxel and carboplatin utilising genome-wide screening in haploid human embryonic stem cells. Cell Prolif 2024:e13771. [PMID: 39523021 DOI: 10.1111/cpr.13771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Taxanes and platinum molecules, specifically paclitaxel and carboplatin, are widely used anticancer drugs that induce cell death and serve as first-line chemotherapy for various cancer types. Despite the efficient effect of both drugs on cancer cell proliferation, many tumours have innate resistance against paclitaxel and carboplatin, which leads to inefficient treatment and poor survival rates. Haploid human embryonic stem cells (hESCs) are a novel and robust platform for genetic screening. To gain a comprehensive view of genes that affect or regulate paclitaxel and carboplatin resistance, genome-wide loss-of-function screens in haploid hESCs were performed. Both paclitaxel and carboplatin screens have yielded selected plausible gene lists and pathways relevant to resistance prediction. The effects of mutations in selected genes on the resistance to the drugs were demonstrated. Based on the results, an algorithm that can predict resistance to paclitaxel or carboplatin was developed. Applying the algorithm to the DNA mutation profile of patients' tumours enabled the separation of sensitive versus resistant patients, thus, providing a prediction tool. As the anticancer drugs arsenal can offer alternatives in case of resistance to either paclitaxel or carboplatin, an early prediction can provide a significant advantage and should improve treatment. The algorithm assists this unmet need and helps predict whether a patient will respond to the treatment and may have an immediate clinically actionable application.
Collapse
Affiliation(s)
| | - Emanuel Segal
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem, Israel
- Hadassah Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | | | - Tamar Golan-Lev
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem, Israel
| | - Benjamin E Reubinoff
- Hadassah Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Adi Turjeman
- The Center for Genomic Technologies, The Hebrew University, Jerusalem, Israel
| | - Ofra Yanuka
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem, Israel
| | | | | | - Nissim Benvenisty
- NewStem LTD, Jerusalem, Israel
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
10
|
Thangavelu L, Altamimi ASA, Ghaboura N, Babu MA, Roopashree R, Sharma P, Pal P, Choudhary C, Prasad GVS, Sinha A, Balaraman AK, Rawat S. Targeting the p53-p21 axis in liver cancer: Linking cellular senescence to tumor suppression and progression. Pathol Res Pract 2024; 263:155652. [PMID: 39437639 DOI: 10.1016/j.prp.2024.155652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
Liver cancer is a major health epidemic worldwide, mainly due to its high mortality rates and limited treatment options. The association of cellular senescence to tumorigenesis and the cancer hallmarks remains a subject of interest in cancer biology. The p53-p21 signalling axis is an important regulator in restoring the cell's balance by supporting tumor suppression and tumorigenesis in liver cancer. We review the novel molecular mechanisms that p53 and its downstream effector, p21, employ to induce cellular senescence, making it last longer, and halt the proliferation of damaged hepatocytes to become tumorous cells. We also examine how dysregulation of this pathway contributes to HCC pathogenesis, proliferation, survival, acquired resistance to apoptosis, and increased invasiveness. Furthermore, we comprehensively describe the molecular cross-talk between the p53-p21 signalling axis and major cell cycle signalling pathways, including Wnt/β-catenin, PI3K/Akt, and TGF-β in liver cancer and provide an overview of promising candidates for chemoprevention and future therapeutic strategies. This review article explores the roles of the p53-p21 pathway in liver cancer, examining its function in promoting cellular senescence under normal conditions and its potential role in cancer progression. It also highlights novel therapeutic drugs and drug targets within the pathway and discusses the implications for treatment strategies and prognosis in liver cancer.
Collapse
Affiliation(s)
- Lakshmi Thangavelu
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Nehmat Ghaboura
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA UNIVERSITY, Mathura, UP 281406, India.
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Pawan Sharma
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Pusparghya Pal
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Chhavi Choudhary
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, Punjab 140307, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - Sushama Rawat
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| |
Collapse
|
11
|
Y KN, Arjunan A, Maigandan D, Dharmarajan A, Perumalsamy LR. Advances and challenges in therapeutic resistant biomarkers of neuroblastoma: A comprehensive review. Biochim Biophys Acta Rev Cancer 2024; 1879:189222. [PMID: 39577750 DOI: 10.1016/j.bbcan.2024.189222] [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: 06/06/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Therapeutic resistance is one of the significant challenges in Neuroblastoma. Owing to its molecular diversity, the therapeutic resistance mechanisms of Neuroblastoma are highly complicated. The traditional chemo and radio therapeutics fail to provide adequate solutions to the treatment resistance, demanding in-depth research to improvise the existing prognostic and therapeutic regimens. To address this knowledge gap, several investigations are being employed, such as unravelling the molecular signalling mechanisms involved in drug resistance at genomics and proteomics levels, development of biomarkers for assessing the therapeutic success, development of novel drug targets for cancer stem cells, targeted immunotherapy and combination therapies. This review collates the ongoing research efforts to address the challenges faced in Neuroblastoma treatment resistance and uncovers the importance of transitioning biomarker discoveries into clinical practice.
Collapse
Affiliation(s)
- Krithicaa Narayanaa Y
- Department of Biomedical Sciences, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India.
| | - Amrutha Arjunan
- Department of Biomedical Sciences, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India.
| | - Devi Maigandan
- Department of Biomedical Sciences, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India.
| | - Arun Dharmarajan
- Sri Ramachandra Faculty of Clinical Research, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India; Curtin Health Innovation Research Institute, Curtin University, Bentley, 6102 Perth, Western Australia, Australia; Curtin Medical School, Curtin University, Perth, Western Australia, Australia; School of Human Sciences, The University of Western Australia, Nedlands, Western Australia, Australia.
| | - Lakshmi R Perumalsamy
- Department of Biomedical Sciences, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India.
| |
Collapse
|
12
|
Kim MK, Cho IR, Kim Y, Choi JH, Jung K, Kim J, Kim S, Yun H, Yoon J, Oh DY, Kim K, Lee SH. Prognostic value of the TP53 mutation in patients with pancreatic ductal adenocarcinoma receiving FOLFIRINOX. Ther Adv Med Oncol 2024; 16:17588359241290482. [PMID: 39449732 PMCID: PMC11500227 DOI: 10.1177/17588359241290482] [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: 04/25/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Background KRAS, TP53, CDKN2A, and SMAD4 have been the main driver mutations in pancreatic ductal adenocarcinoma (PDAC). Studies on the clinical significance and treatment response to 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) regimen in terms of the presence of these mutations remain inconclusive. Objectives This study aimed to compare the survival outcome and response to FOLFIRINOX chemotherapy based on the presence of four driver mutation genes. Design A multi-center retrospective study conducted at two tertiary medical centers. Methods This study analyzed PDAC patients who were treated with FOLFIRINOX chemotherapy as the initial treatment. Tumor specimens were analyzed by a targeted next-generation sequencing platform at two tertiary referral hospitals from January 2016 to March 2022. Patients' demographics, survival outcomes, and chemotherapeutic response were investigated and compared according to the presence of driver mutations. Results The analysis included 100 patients. KRAS mutation was identified in 92 (92.0%) patients, followed by TP53, CDKN2A, and SMAD4 in 63 (63.0%), 18 (18.0%), and 17 (17.0%) patients, respectively. The TP53 wild-type group demonstrated longer overall survival (OS) than the TP53 mutated group (median OS: 29 vs 19 months, p = 0.03), and TP53 served as a prognostic factor for survival (hazard ratio = 1.74, 95% confidence interval: 1.00-3.00, p = 0.048). The difference in OS according to TP53 mutation was intensified in localized pancreatic adenocarcinoma (37 vs 19 months, p = 0.01). The TP53 wild-type group demonstrated a higher objective response rate to FOLFIRINOX chemotherapy than the TP53 mutation group in localized pancreatic adenocarcinoma (50.0% vs 17.6%, p = 0.024). Conclusion PDAC patients with wild-type TP53 demonstrated longer OS than those with TP53 mutation, and this trend was intensified in patients with localized disease. This result may be due to an impaired response to FOLFIRINOX chemotherapy in patients with TP53 mutation.
Collapse
Affiliation(s)
- Min Kyu Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - In Rae Cho
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yooeun Kim
- Interdisciplinary Program in Bioinformatics, College of Natural Science, Seoul National University, Seoul, Republic of Korea
| | - Jin Ho Choi
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kwangrok Jung
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam-si, Republic of Korea
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam-si, Republic of Korea
| | - Sheehyun Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hongseok Yun
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jeesun Yoon
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Do-Youn Oh
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kwangsoo Kim
- Department of Transdisciplinary Medicine, Institute of Convergence Medicine with Innovative Technology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sang Hyub Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| |
Collapse
|
13
|
Chen SJ, Tsai CC, Lin SR, Lee MH, Huang SS, Zeng HY, Wang LH, Chiang MF, Sheu HM, Chang NS. Dissociation of the nuclear WWOX/TRAF2 switch renders UV/cold shock-mediated nuclear bubbling cell death at low temperatures. Cell Commun Signal 2024; 22:505. [PMID: 39420317 PMCID: PMC11487720 DOI: 10.1186/s12964-024-01866-6] [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: 06/02/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Normal cells express functional tumor suppressor WW domain-containing oxidoreductase (WWOX), designated WWOXf. UV irradiation induces WWOXf cells to undergo bubbling cell death (BCD) - an event due to the accumulation of nuclear nitric oxide (NO) gas that forcefully pushes the nuclear and cell membranes to form one or two bubbles at room temperature (22 °C) and below. In contrast, when WWOX-deficient or -dysfunctional (WWOXd) cells are exposed to UV and/or cold shock, the cells undergo nuclear pop-out explosion death (POD). We aimed to determine the morphological and biochemical changes in WWOXf cells during BCD versus apoptosis. METHODS WWOXf and WWOXd cells were exposed to UV followed by measuring BCD or POD by time-lapse microscopy and/or time-lapse holographic microscopy at 4, 22, or 37 °C to visualize morphological changes. Live cell stains were used to measure the kinetics of nitric oxide (NO) production and Ca2+ influx. Extent of cell death was measured by uptake of propidium iodide and by internucleosomal DNA fragmentation using agarose gel electrophoresis. RESULTS WWOXf cells were exposed to UV and then cold shock, or cold shock and then UV, and cultured at 4, 10, and 22 °C, respectively. Initially, UV induced calcium influx and NO production, which led to nuclear bubbling and final death. Cold shock pretreatment completely suppressed UV-mediated bubbling at 37 °C, so the UV/cold shock-treated cells underwent apoptosis. Without cold shock, UV only induced bubbling at all temperatures, whereas the efficiency of bubbling at 37 °C was reduced by greater than 50%. Morphologically, the WWOXf cell height or thickness was significantly increased during cell division or apoptosis, but the event did not occur in BCD. In comparison, when WWOXd cancer cells received UV or UV/cold shock, these cells underwent NO-independent POD. UV/cold shock effectively downregulated the expression of many proteins such as the housekeeping α-tubulin (> 70%) and β-actin (< 50%), and cortactin (> 70%) in WWOXf COS7 cells. UV/cold shock induced relocation of α-tubulin to the nucleus and nuclear bubbles in damaged cells. UV induced co-translocation of the WWOX/TRAF2 complex to the nuclei, in which the prosurvival TRAF2 blocked the proapoptotic WWOX via its zinc finger domain. Without WWOX, TRAF2 did not relocate to the nuclei. Cold shock caused the dissociation of the WWOX/TRAF2 complex in the nucleus needed for BCD. In contrast, the formation of the WWOX/TRAF2 complex, plus p53, was strengthened at 37 °C required for apoptosis. CONCLUSIONS The temperature-sensitive nuclear WWOX/TRAF2 complex acts as a molecular switch, whose dissociation favors BCD at low temperatures, and the association supports apoptosis at 37 °C in UV-treated WWOXf cells.
Collapse
Affiliation(s)
- Szu-Jung Chen
- Institute of Molecular Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Cheng-Chang Tsai
- Institute of Molecular Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Sing-Ru Lin
- Institute of Molecular Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Ming-Hui Lee
- Institute of Molecular Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Shenq-Shyang Huang
- Institute of Molecular Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Han-Yan Zeng
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Lu-Hai Wang
- Chinese Medicine Research Center, Institute of Integrated Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Ming-Fu Chiang
- Department of Neurosurgery, Fu Jen Catholic University Hospital, Taipei, 24352, Taiwan.
| | - Hamm-Ming Sheu
- Department of Dermatology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
| | - Nan-Shan Chang
- Institute of Molecular Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
14
|
Stojanović Gužvić N, Lüke F, Treitschke S, Coluccio A, Hoffmann M, Feliciello G, Varadarajan AR, Lu X, Weidele K, Botteron C, Materna-Reichelt S, Keil F, Evert K, Weber F, Schamberger T, Althammer M, Grosse J, Hellwig D, Schulz C, Seitz S, Ugocsai P, Schlenska-Lange A, Mayr R, Kaiser U, Dietmaier W, Polzer B, Warfsmann J, Honarnejad K, Pukrop T, Heudobler D, Klein CA, Werno C. Cellular liquid biopsy provides unique chances for disease monitoring, preclinical model generation and therapy adjustment in rare salivary gland cancer patients. Mol Oncol 2024. [PMID: 39367702 DOI: 10.1002/1878-0261.13741] [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: 01/30/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 10/06/2024] Open
Abstract
While cell-free liquid biopsy (cfLB) approaches provide simple and inexpensive disease monitoring, cell-based liquid biopsy (cLB) may enable additional molecular genetic assessment of systemic disease heterogeneity and preclinical model development. We investigated 71 blood samples of 62 patients with various advanced cancer types and subjected enriched circulating tumor cells (CTCs) to organoid culture conditions. CTC-derived tumoroid models were characterized by DNA/RNA sequencing and immunohistochemistry, as well as functional drug testing. Results were linked to molecular features of primary tumors, metastases, and CTCs; CTC enumeration was linked to disease progression. Of 52 samples with positive CTC counts (≥1) from eight different cancer types, only CTCs from two salivary gland cancer (SGC) patients formed tumoroid cultures (P = 0.0005). Longitudinal CTC enumeration of one SGC patient closely reflected disease progression during treatment and revealed metastatic relapse earlier than clinical imaging. Multiomics analysis and functional in vitro drug testing identified potential resistance mechanisms and drug vulnerabilities. We conclude that cLB might add a functional dimension (to the genetic approaches) in the personalized management of rare, difficult-to-treat cancers such as SGC.
Collapse
Affiliation(s)
| | - Florian Lüke
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Steffi Treitschke
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | - Andrea Coluccio
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | - Martin Hoffmann
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | | | | | - Xin Lu
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | - Kathrin Weidele
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | - Catherine Botteron
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | | | - Felix Keil
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Insitute for Pathology, University of Regensburg, Germany
| | - Katja Evert
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Insitute for Pathology, University of Regensburg, Germany
| | - Florian Weber
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Insitute for Pathology, University of Regensburg, Germany
| | - Thomas Schamberger
- Experimental Medicine and Therapy Research, University of Regensburg, Germany
| | - Michael Althammer
- Experimental Medicine and Therapy Research, University of Regensburg, Germany
| | - Jirka Grosse
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Department of Nuclear Medicine, University Hospital Regensburg, Germany
| | - Dirk Hellwig
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Department of Nuclear Medicine, University Hospital Regensburg, Germany
| | - Christian Schulz
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Department of Internal Medicine II, University Hospital Regensburg, Germany
| | - Stephan Seitz
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Department of Obstetrics and Gynecology, University Hospital Regensburg, Germany
| | - Peter Ugocsai
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Department of Obstetrics and Gynecology, University Hospital Regensburg, Germany
| | - Anke Schlenska-Lange
- Department of Oncology and Hematology, Hospital Barmherzige Brüder, Regensburg, Germany
| | - Roman Mayr
- Department of Urology, Caritas St. Josef Medical Center, University of Regensburg, Germany
| | - Ulrich Kaiser
- Department of Internal Medicine III, University Hospital Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | | | - Bernhard Polzer
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | - Jens Warfsmann
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | - Kamran Honarnejad
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | - Tobias Pukrop
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Daniel Heudobler
- Department of Internal Medicine III, University Hospital Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Christoph A Klein
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Experimental Medicine and Therapy Research, University of Regensburg, Germany
| | - Christian Werno
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| |
Collapse
|
15
|
Zamanian MY, Taheri N, Ramadan MF, Mustafa YF, Alkhayyat S, Sergeevna KN, Alsaab HO, Hjazi A, Molavi Vasei F, Daneshvar S. A comprehensive view on the fisetin impact on colorectal cancer in animal models: Focusing on cellular and molecular mechanisms. Animal Model Exp Med 2024; 7:591-605. [PMID: 39136058 PMCID: PMC11528395 DOI: 10.1002/ame2.12476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/21/2024] [Accepted: 07/09/2024] [Indexed: 11/02/2024] Open
Abstract
Flavonoids, including fisetin, have been linked to a reduced risk of colorectal cancer (CRC) and have potential therapeutic applications for the condition. Fisetin, a natural flavonoid found in various fruits and vegetables, has shown promise in managing CRC due to its diverse biological activities. It has been found to influence key cell signaling pathways related to inflammation, angiogenesis, apoptosis, and transcription factors. The results of this study demonstrate that fisetin induces colon cancer cell apoptosis through multiple mechanisms. It impacts the p53 pathway, leading to increased levels of p53 and decreased levels of murine double minute 2, contributing to apoptosis induction. Fisetin also triggers the release of important components in the apoptotic process, such as second mitochondria-derived activator of caspase/direct inhibitor of apoptosis-binding protein with low pI and cytochrome c. Furthermore, fisetin inhibits the cyclooxygenase-2 and wingless-related integration site (Wnt)/epidermal growth factor receptor/nuclear factor kappa B signaling pathways, reducing Wnt target gene expression and hindering colony formation. It achieves this by regulating the activities of cyclin-dependent kinase 2 and cyclin-dependent kinase 4, reducing retinoblastoma protein phosphorylation, decreasing cyclin E levels, and increasing p21 levels, ultimately influencing E2 promoter binding factor 1 and cell division cycle 2 (CDC2) protein levels. Additionally, fisetin exhibits various effects on CRC cells, including inhibiting the phosphorylation of Y-box binding protein 1 and ribosomal S6 kinase, promoting the phosphorylation of extracellular signal-regulated kinase 1/2, and disrupting the repair process of DNA double-strand breaks. Moreover, fisetin serves as an adjunct therapy for the prevention and treatment of phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (PIK3CA)-mutant CRC, resulting in a reduction in phosphatidylinositol-3 kinase (PI3K) expression, Ak strain transforming phosphorylation, mTOR activity, and downstream target proteins in CRC cells with a PIK3CA mutation. These findings highlight the multifaceted potential of fisetin in managing CRC and position it as a promising candidate for future therapy development.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Department of Physiology, School of MedicineHamadan University of Medical SciencesHamadanIran
- Department of Pharmacology and Toxicology, School of PharmacyHamadan University of Medical SciencesHamadanIran
| | - Niloofar Taheri
- School of MedicineShahroud University of Medical SciencesShahroudIran
| | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical ChemistryCollege of Pharmacy, University of MosulMosulIraq
| | | | - Klunko Nataliya Sergeevna
- Department of Training of Scientific and Scientific‐Pedagogical PersonnelRussian New UniversityMoscowRussian Federation
| | - Hashem O. Alsaab
- Department of Pharmaceutics and Pharmaceutical TechnologyTaif UniversityTaifSaudi Arabia
| | - Ahmed Hjazi
- Department of Medical LaboratoryCollege of Applied Medical Sciences, Prince Sattam bin Abdulaziz UniversityAl‐KharjSaudi Arabia
| | - Farnoosh Molavi Vasei
- Department of Clinical Biochemistry, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| | - Siamak Daneshvar
- Department of Surgery, School of MedicineHamadan University of Medical SciencesHamadanIran
| |
Collapse
|
16
|
Zhang X, Lin Y, He D, Sun M, Xu L, Chang Z, Liu Z, Li B. 18F-Fluoro-2-Deoxyglucose Positron Emission Tomography/Computed Tomography Measures of Spatial Heterogeneity for Predicting Platinum Resistance of High-Grade Serous Ovarian Cancer. Cancer Med 2024; 13:e70287. [PMID: 39435561 PMCID: PMC11494247 DOI: 10.1002/cam4.70287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/02/2024] [Accepted: 09/22/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND The purpose of this study is to construct models for predicting platinum resistance in high-grade serous ovarian cancer (HGSOC) derived from quantitative spatial heterogeneity indicators obtained from 18F-FDG PET/CT images. METHODS A retrospective study was conducted on patients diagnosed with HGSOC. Quantitative indicators of spatial heterogeneity were generated using conventional features and Haralick texture features from both CT and PET images. Three groups of predictive models (conventional, heterogeneity, and integrated) were built. Each group's optimal model was the one with the highest area under curve (AUC). Postoperative immunohistochemical staining for Ki-67 and p53 was conducted. The correlation between the heterogeneity indicators and scores for Ki-67 and p53 was assessed by Spearman's correlation coefficient (ρ). RESULTS A total of 286 patients (54.6 ± 9.3 years) were enrolled. And 107 spatial heterogeneity indicators were extracted. The optimal models for each group were obtained using the Gradient Boosting Machine (GBM) algorithm. There was an AUC of 0.790 (95% CI: 0.696, 0.885) in the conventional model for the validation set, and an AUC of 0.904 (95% CI: 0.842, 0.966) in the heterogeneity model for the validation set. The integrated model achieved the highest predictive performance, with an AUC value of 0.928 (95% CI: 0.872, 0.984) for the validation set. Spearman's correlation showed that HU_Kurtosis had the strongest correlation with p53 scores with ρ = 0.718, while cluster site entropy had the strongest correlation with Ki-67 scores with ρ = 0.753. CONCLUSIONS Adding quantitative spatial heterogeneity indicators derived from PET/CT images can improve the prediction of platinum resistance in patients with HGSOC. Spatial heterogeneity indicators were related to Ki-67 and p53 scores.
Collapse
Affiliation(s)
- Xin Zhang
- Department of General SurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningPeople's Republic of China
| | - Yuhe Lin
- Department of OncologyShengjing Hospital of China Medical UniversityShenyangLiaoningPeople's Republic of China
| | - Dianning He
- School of Health ManagementChina Medical UniversityShenyangLiaoningPeople's Republic of China
| | - Mingli Sun
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangLiaoningPeople's Republic of China
| | - Lanlan Xu
- Department of RadiologyShengjing Hospital of China Medical UniversityShenyangLiaoningPeople's Republic of China
| | - Zhihui Chang
- Department of RadiologyShengjing Hospital of China Medical UniversityShenyangLiaoningPeople's Republic of China
| | - Zhaoyu Liu
- Department of RadiologyShengjing Hospital of China Medical UniversityShenyangLiaoningPeople's Republic of China
| | - Beibei Li
- Department of RadiologyShengjing Hospital of China Medical UniversityShenyangLiaoningPeople's Republic of China
| |
Collapse
|
17
|
Thepmalee C, Jenkham P, Ramwarungkura B, Suwannasom N, Khoothiam K, Thephinlap C, Sawasdee N, Panya A, Yenchitsomanus PT. Enhancing cancer immunotherapy using cordycepin and Cordyceps militaris extract to sensitize cancer cells and modulate immune responses. Sci Rep 2024; 14:21907. [PMID: 39300166 DOI: 10.1038/s41598-024-72833-x] [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: 05/21/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
Integrating immunotherapy with natural compounds holds promise in enhancing the immune system's ability to eliminate cancer cells. Cordyceps militaris, a traditional Chinese medicine, emerges as a promising candidate in this regard. This study investigates the effects of cordycepin and C. militaris ethanolic extract (Cm-EE) on sensitizing cancer cells and regulating immune responses against breast cancer (BC) and hepatocellular carcinoma (HCC) cells. Cordycepin, pentostatin and adenosine were identified in Cm-EE. Cordycepin treatment decreased HLA-ABC-positive cells in pre-treated cancer cells, while Cm-EE increased NKG2D ligand and death receptor expression. Additionally, cordycepin enhanced NKG2D receptor and death ligand expression on CD3-negative effector immune cells, particularly on natural killer (NK) cells, while Cm-EE pre-treatment stimulated IL-2, IL-6, and IL-10 production. Co-culturing cancer cells with effector immune cells during cordycepin or Cm-EE incubation resulted in elevated cancer cell death. These findings highlight the potential of cordycepin and Cm-EE in improving the efficacy of cancer immunotherapy for BC and HCC.
Collapse
Affiliation(s)
- Chutamas Thepmalee
- Unit of Excellence on Research and Development of Cancer Therapy, University of Phayao, Phayao, Thailand
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao, Thailand
| | - Phanitaporn Jenkham
- Unit of Excellence on Research and Development of Cancer Therapy, University of Phayao, Phayao, Thailand
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao, Thailand
| | - Boonyanuch Ramwarungkura
- Division of Molecular Medicine, Research Department, Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE‑CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nittiya Suwannasom
- Unit of Excellence on Research and Development of Cancer Therapy, University of Phayao, Phayao, Thailand
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao, Thailand
| | - Krissana Khoothiam
- Unit of Excellence on Research and Development of Cancer Therapy, University of Phayao, Phayao, Thailand
- Division of Microbiology, School of Medical Sciences, University of Phayao, Phayao, Thailand
| | - Chonthida Thephinlap
- Unit of Excellence on Research and Development of Cancer Therapy, University of Phayao, Phayao, Thailand
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao, Thailand
| | - Nunghathai Sawasdee
- Division of Molecular Medicine, Research Department, Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE‑CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Aussara Panya
- Cell Engineering for Cancer Therapy Research Group, Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai, Thailand
| | - Pa-Thai Yenchitsomanus
- Division of Molecular Medicine, Research Department, Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE‑CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
18
|
Sue SH, Tseng WC, Wu ZS, Huang SM, Chen JL, Wu ZF, Lai HC. The synergistic mechanisms of propofol with cisplatin or doxorubicin in human ovarian cancer cells. J Ovarian Res 2024; 17:187. [PMID: 39272193 PMCID: PMC11401282 DOI: 10.1186/s13048-024-01509-x] [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: 01/17/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Most ovarian cancer cases are diagnosed at an advanced stage, leading to poor outcomes and a relatively low 5-year survival rate. While tumor resection in the early stages can be highly effective, recurrence following primary treatment remains a significant cause of mortality. Propofol is a commonly used intravenous anesthetic agent in cancer resection surgery. Previous research has shown that propofol anesthesia was associated with improved survival in patients undergoing elective surgery for epithelial ovarian cancer. However, the underlying antitumor mechanisms are not yet fully understood. METHODS This study aimed to uncover the antitumor properties of propofol alone and combined with cisplatin or doxorubicin, in human SKOV3 and OVCAR3 ovarian cancer cells. We applied flowcytometry analysis for mitochondrial membrane potential, apoptosis, and autophagy, colony formation, migration, and western blotting analysis. RESULTS Given that chemotherapy is a primary clinical approach for managing advanced and recurrent ovarian cancer, it is essential to address the limitations of current chemotherapy, particularly in the use of cisplatin and doxorubicin, which are often constrained by their side effects and the development of resistance. First of all, propofol acted synergistically with cisplatin and doxorubicin in SKOV3 cells. Moreover, our data further showed that propofol suppressed colony formation, disrupted mitochondrial membrane potential, and induced apoptosis and autophagy in SKOV3 and OVCAR3 cells. Finally, the effects of combined propofol with cisplatin or doxorubicin on mitochondrial membrane potential, apoptosis, autophagy, and epithelial-mesenchymal transition were different in SKOV3 and OVCAR3 cells, depending on the p53 status. CONCLUSION In summary, repurposing propofol could provide novel insights into the existing chemotherapy strategies for ovarian cancer. It holds promise for overcoming resistance to cisplatin or doxorubicin and may potentially reduce the required chemotherapy dosages and associated side effects, thus improving treatment outcomes.
Collapse
Affiliation(s)
- Sung-How Sue
- Department of Surgery, Taipei City Hospital Renai Branch, Taipei City, 106, Taiwan, Republic of China
| | - Wei-Cheng Tseng
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114, Taiwan, Republic of China
| | - Zih-Syuan Wu
- Institute of Life Sciences, National Defense Medical Center, Taipei City, 114, Taiwan, Republic of China
| | - Shih-Ming Huang
- Institute of Life Sciences, National Defense Medical Center, Taipei City, 114, Taiwan, Republic of China
- Department of Biochemistry, National Defense Medical Center, Taipei City, 114, Taiwan, Republic of China
| | - Jia-Lin Chen
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114, Taiwan, Republic of China.
| | - Zhi-Fu Wu
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City 807, Taiwan, Republic of China.
- Department of Anesthesiology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City 807, Taiwan, Republic of China.
- Center for Regional Anesthesia and Pain Medicine, Wan Fang Hospital, Taipei Medical University, Taipei City 116, Taiwan, Taiwan, Republic of China.
| | - Hou-Chuan Lai
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114, Taiwan, Republic of China.
| |
Collapse
|
19
|
Khosravani H, Ataee Disfani R, Farhadi B, Tohidian M, Garrosi L, Shirvani P, Zabihi MR, Akhoondian M, Norouzkhani N, Farzan R. Esophageal chemical burns as a risk factor for esophageal malignancies: in-silico analyses - experimental research. Ann Med Surg (Lond) 2024; 86:5170-5178. [PMID: 39239040 PMCID: PMC11374246 DOI: 10.1097/ms9.0000000000002317] [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: 03/20/2024] [Accepted: 06/17/2024] [Indexed: 09/07/2024] Open
Abstract
Introduction Esophageal chemical burns often occur through accidental or intentional oral consumption of chemical agents and lead to severe complications such as esophageal stricture, acute perforation, and even death. Esophageal squamous cell carcinoma is a squamous epithelium tumor that lines the normal esophagus. Additionally, adenocarcinomas are tumors located at the interface between the distal esophagus and the proximal gastric and divided into esophageal adenocarcinoma and gastric-cardia adenocarcinoma. Various conditions, such as chemical burns, are considered risk factors in the disease's pathogenesis. In the in-silico study, the authors aim to present the relationship between chemical burns and esophageal cancer by analyzing bioinformatics genetic data. Methods The proper gene set was extracted using the 'GEO' database. The string web tool was utilized to form the gene-interaction network. Gephi and Cytoscape software were applied to achieve network analysis. Results According to in-silico data, 26 genes, including NCAPH, DLGAP5, CCNB1, KIF11, KIAA0101, CDCA5, BIRC5, NUF2, BUB1B, RRM2, TTK, CDC20, NUSAP1, CCNB2, CCNA2, MELK, TPX2, PRC1, KIF4A, CENPF, TOP2A, CDK1, ASPM, CEP55, BUB1, KIF20A were extracted that can be regarded as the most critical shared genes between chemical burns and esophageal cancer. Conclusion In sum, esophageal chemical burns can be related to the occurrence of esophageal cancer. Moreover, esophageal chemical burn is an external factor that upregulates present genes and can be regarded as a worsening prognosis or risk factor for esophageal cancer.
Collapse
Affiliation(s)
| | - Reza Ataee Disfani
- Student Research Committee, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Bahar Farhadi
- School of Medicine, Islamic Azad University, Mashhad Branch, Mashhad, Iran
| | - Mobina Tohidian
- Department of Anatomy and Cell Biology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Lida Garrosi
- Department of Obstetrics and Gynecology, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Proushat Shirvani
- School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Reza Zabihi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Akhoondian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Narges Norouzkhani
- Department of Medical Informatics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramyar Farzan
- Department of Plastic & Reconstructive Surgery, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
20
|
McCallum N, Najlah M. The Anticancer Activity of Monosaccharides: Perspectives and Outlooks. Cancers (Basel) 2024; 16:2775. [PMID: 39199548 PMCID: PMC11353049 DOI: 10.3390/cancers16162775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 07/27/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024] Open
Abstract
A major hallmark of cancer is the reprogramming of cellular metabolism from oxidative phosphorylation (OXPHOS) to glycolysis, a phenomenon known as the Warburg effect. To sustain high rates of glycolysis, cancer cells overexpress GLUT transporters and glycolytic enzymes, allowing for the enhanced uptake and consumption of glucose. The Warburg effect may be exploited in the treatment of cancer; certain epimers and derivatives of glucose can enter cancer cells and inhibit glycolytic enzymes, stunting metabolism and causing cell death. These include common dietary monosaccharides (ᴅ-mannose, ᴅ-galactose, ᴅ-glucosamine, ʟ-fucose), as well as some rare monosaccharides (xylitol, ᴅ-allose, ʟ-sorbose, ʟ-rhamnose). This article reviews the literature on these sugars in in vitro and in vivo models of cancer, discussing their mechanisms of cytotoxicity. In addition to this, the anticancer potential of some synthetically modified monosaccharides, such as 2-deoxy-ᴅ-glucose and its acetylated and halogenated derivatives, is reviewed. Further, this article reviews how certain monosaccharides can be used in combination with anticancer drugs to potentiate conventional chemotherapies and to help overcome chemoresistance. Finally, the limitations of administering two separate agents, a sugar and a chemotherapeutic drug, are discussed. The potential of the glycoconjugation of classical or repurposed chemotherapy drugs as a solution to these limitations is reviewed.
Collapse
Affiliation(s)
| | - Mohammad Najlah
- Pharmaceutical Research Group, School of Allied Health, Faculty of Health, Education, Medicine and Social Care, Anglia Ruskin University, Bishops Hall Lane, Chelmsford CM1 1SQ, UK;
| |
Collapse
|
21
|
Visuddho V, Halim P, Helen H, Muhar AM, Iqhrammullah M, Mayulu N, Surya R, Tjandrawinata RR, Ribeiro RIMA, Tallei TE, Taslim NA, Kim B, Syahputra RA, Nurkolis F. Modulation of Apoptotic, Cell Cycle, DNA Repair, and Senescence Pathways by Marine Algae Peptides in Cancer Therapy. Mar Drugs 2024; 22:338. [PMID: 39195454 DOI: 10.3390/md22080338] [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: 05/25/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
Marine algae, encompassing both macroalgae and microalgae, have emerged as a promising and prolific source of bioactive compounds with potent anticancer properties. Despite their significant therapeutic potential, the clinical application of these peptides is hindered by challenges such as poor bioavailability and susceptibility to enzymatic degradation. To overcome these limitations, innovative delivery systems, particularly nanocarriers, have been explored. Nanocarriers, including liposomes, nanoparticles, and micelles, have demonstrated remarkable efficacy in enhancing the stability, solubility, and bioavailability of marine algal peptides, ensuring controlled release and prolonged therapeutic effects. Marine algal peptides encapsulated in nanocarriers significantly enhance bioavailability, ensuring more efficient absorption and utilization in the body. Preclinical studies have shown promising results, indicating that nanocarrier-based delivery systems can significantly improve the pharmacokinetic profiles and therapeutic outcomes of marine algal peptides. This review delves into the diverse anticancer mechanisms of marine algal peptides, which include inducing apoptosis, disrupting cell cycle progression, and inhibiting angiogenesis. Further research focused on optimizing nanocarrier formulations, conducting comprehensive clinical trials, and continued exploration of marine algal peptides holds great promise for developing innovative, effective, and sustainable cancer therapies.
Collapse
Affiliation(s)
- Visuddho Visuddho
- Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Princella Halim
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Helen Helen
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Adi Muradi Muhar
- Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Muhammad Iqhrammullah
- Postgraduate Program of Public Health, Universitas Muhammadiyah Aceh, Banda Aceh 23123, Indonesia
| | - Nelly Mayulu
- Department of Nutrition, Faculty of Health Science, Muhammadiyah Manado University, Manado 95249, Indonesia
| | - Reggie Surya
- Department of Food Technology, Faculty of Engineering, Bina Nusantara University, Jakarta 11480, Indonesia
| | - Raymond Rubianto Tjandrawinata
- Department of Biotechnology, Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, Jakarta 12930, Indonesia
| | | | - Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado 95115, Indonesia
| | - Nurpudji Astuti Taslim
- Division of Clinical Nutrition, Department of Nutrition, Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Fahrul Nurkolis
- Department of Biological Sciences, Faculty of Sciences and Technology, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta 55281, Indonesia
| |
Collapse
|
22
|
Drif AI, Yücer R, Damiescu R, Ali NT, Abu Hagar TH, Avula B, Khan IA, Efferth T. Anti-Inflammatory and Cancer-Preventive Potential of Chamomile ( Matricaria chamomilla L.): A Comprehensive In Silico and In Vitro Study. Biomedicines 2024; 12:1484. [PMID: 39062057 PMCID: PMC11275008 DOI: 10.3390/biomedicines12071484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND AND AIM Chamomile tea, renowned for its exquisite taste, has been appreciated for centuries not only for its flavor but also for its myriad health benefits. In this study, we investigated the preventive potential of chamomile (Matricaria chamomilla L.) towards cancer by focusing on its anti-inflammatory activity. METHODS AND RESULTS A virtual drug screening of 212 phytochemicals from chamomile revealed β-amyrin, β-eudesmol, β-sitosterol, apigenin, daucosterol, and myricetin as potent NF-κB inhibitors. The in silico results were verified through microscale thermophoresis, reporter cell line experiments, and flow cytometric determination of reactive oxygen species and mitochondrial membrane potential. An oncobiogram generated through comparison of 91 anticancer agents with known modes of action using the NCI tumor cell line panel revealed significant relationships of cytotoxic chamomile compounds, lupeol, and quercetin to microtubule inhibitors. This hypothesis was verified by confocal microscopy using α-tubulin-GFP-transfected U2OS cells and molecular docking of lupeol and quercetin to tubulins. Both compounds induced G2/M cell cycle arrest and necrosis rather than apoptosis. Interestingly, lupeol and quercetin were not involved in major mechanisms of resistance to established anticancer drugs (ABC transporters, TP53, or EGFR). Performing hierarchical cluster analyses of proteomic expression data of the NCI cell line panel identified two sets of 40 proteins determining sensitivity and resistance to lupeol and quercetin, further pointing to the multi-specific nature of chamomile compounds. Furthermore, lupeol, quercetin, and β-amyrin inhibited the mRNA expression of the proinflammatory cytokines IL-1β and IL6 in NF-κB reporter cells (HEK-Blue Null1). Moreover, Kaplan-Meier-based survival analyses with NF-κB as the target protein of these compounds were performed by mining the TCGA-based KM-Plotter repository with 7489 cancer patients. Renal clear cell carcinomas (grade 3, low mutational rate, low neoantigen load) were significantly associated with shorter survival of patients, indicating that these subgroups of tumors might benefit from NF-κB inhibition by chamomile compounds. CONCLUSION This study revealed the potential of chamomile, positioning it as a promising preventive agent against inflammation and cancer. Further research and clinical studies are recommended.
Collapse
Affiliation(s)
- Assia I. Drif
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Rümeysa Yücer
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Roxana Damiescu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Nadeen T. Ali
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Tobias H. Abu Hagar
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Bharati Avula
- National Center for Natural Products Research (NCNPR), School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (B.A.); (I.A.K.)
| | - Ikhlas A. Khan
- National Center for Natural Products Research (NCNPR), School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (B.A.); (I.A.K.)
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| |
Collapse
|
23
|
Peng Z, Ahsan N, Yang Z. Proteomics Analysis of Interactions between Drug-Resistant and Drug-Sensitive Cancer Cells: Comparative Studies of Monoculture and Coculture Cell Systems. J Proteome Res 2024; 23:2608-2618. [PMID: 38907724 PMCID: PMC11425778 DOI: 10.1021/acs.jproteome.4c00338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Cell-cell interactions, which allow cells to communicate with each other through molecules in their microenvironment, are critical for the growth, health, and functions of cells. Previous studies show that drug-resistant cells can interact with drug-sensitive cells to elevate their drug resistance level, which is partially responsible for cancer recurrence. Studying protein targets and pathways involved in cell-cell communication provides essential information for fundamental cell biology studies and therapeutics of human diseases. In the current studies, we performed direct coculture and indirect coculture of drug-resistant and drug-sensitive cell lines, aiming to investigate intracellular proteins responsible for cell communication. Comparative studies were carried out using monoculture cells. Shotgun bottom-up proteomics results indicate that the P53 signaling pathway has a strong association with drug resistance mechanisms, and multiple TP53-related proteins were upregulated in both direct and indirect coculture systems. In addition, cell-cell communication pathways, including the phagosome and the HIF-signaling pathway, contribute to both direct and indirect coculture systems. Consequently, AK3 and H3-3A proteins were identified as potential targets for cell-cell interactions that are relevant to drug resistance mechanisms. We propose that the P53 signaling pathway, in which mitochondrial proteins play an important role, is responsible for inducing drug resistance through communication between drug-resistant and drug-sensitive cancer cells.
Collapse
Affiliation(s)
- Zongkai Peng
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Nagib Ahsan
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
- Mass Spectrometry, Proteomics and Metabolomics Core Facility, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Zhibo Yang
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| |
Collapse
|
24
|
Liao W, Zhang R, Chen G, Zhu X, Wu W, Chen Z, Jiang C, Lin Z, Ma L, Yu H. Berberine synergises with ferroptosis inducer sensitizing NSCLC to ferroptosis in p53-dependent SLC7A11-GPX4 pathway. Biomed Pharmacother 2024; 176:116832. [PMID: 38850659 DOI: 10.1016/j.biopha.2024.116832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/21/2024] [Accepted: 05/26/2024] [Indexed: 06/10/2024] Open
Abstract
Berberine (BBR) is a compound derived from Chinese herbal medicine, known for its anticancer properties through multiple signaling pathways. However, whether BBR can inhibit tumor growth by participating in ferroptosis remains unconfirmed. In this study, we demonstrated that berberine synergistically inhibited NSCLC in combination with multiple ferroptosis inducers, and this combination synergistically down-regulated the mRNA and protein expression of SLC7A11, GPX4, and NRF2, resulting in ferroptosis accompanied by significant depletion of GSH, and aberrant accumulation of reactive oxygen species and malondialdehyde. In a lung cancer allograft model, the combination treatment exhibited enhanced anticancer effects compared to using either drug alone. Notably, p53 is critical in determining the ferroptosis sensitivity. We found that the combination treatment did not elicit a synergistic anticancer effect in cells with a p53 mutation or with exogenous expression of mutant p53. These findings provide insight into the mechanism by which combination induces ferroptosis and the regulatory role of p53 in this process. It may guide the development of new strategies for treating NSCLC, offering great medical potential for personal diagnosis and treatment.
Collapse
Affiliation(s)
- Weilin Liao
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao Special Administrative Region of China
| | - Ren Zhang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao Special Administrative Region of China
| | - Geer Chen
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao Special Administrative Region of China
| | - Xiaoyu Zhu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao Special Administrative Region of China
| | - Weiyu Wu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao Special Administrative Region of China
| | - Ziyu Chen
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao Special Administrative Region of China
| | - Chenyu Jiang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao Special Administrative Region of China
| | - Zicong Lin
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao Special Administrative Region of China
| | - Lijuan Ma
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao Special Administrative Region of China
| | - Haijie Yu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao Special Administrative Region of China.
| |
Collapse
|
25
|
Shinhmar S, Schaf J, Lloyd Jones K, Pardo OE, Beesley P, Williams RSB. Developing a Tanshinone IIA Memetic by Targeting MIOS to Regulate mTORC1 and Autophagy in Glioblastoma. Int J Mol Sci 2024; 25:6586. [PMID: 38928292 PMCID: PMC11204349 DOI: 10.3390/ijms25126586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Tanshinone IIA (T2A) is a bioactive compound that provides promise in the treatment of glioblastoma multiforme (GBM), with a range of molecular mechanisms including the inhibition of the mechanistic target of rapamycin complex 1 (mTORC1) and the induction of autophagy. Recently, T2A has been demonstrated to function through sestrin 2 (SESN) to inhibit mTORC1 activity, but its possible impact on autophagy through this pathway has not been investigated. Here, the model system Dictyostelium discoideum and GBM cell lines were employed to investigate the cellular role of T2A in regulating SESN to inhibit mTORC1 and activate autophagy through a GATOR2 component MIOS. In D. discoideum, T2A treatment induced autophagy and inhibited mTORC1 activity, with both effects lost upon the ablation of SESN (sesn-) or MIOS (mios-). We further investigated the targeting of MIOS to reproduce this effect of T2A, where computational analysis identified 25 novel compounds predicted to strongly bind the human MIOS protein, with one compound (MIOS inhibitor 3; Mi3) reducing cell proliferation in two GBM cells. Furthermore, Mi3 specificity was demonstrated through the loss of potency in the D. discoideum mios- cells regarding cell proliferation and the induction of autophagy. In GBM cells, Mi3 treatment also reduced mTORC1 activity and induced autophagy. Thus, a potential T2A mimetic showing the inhibition of mTORC1 and induction of autophagy in GBM cells was identified.
Collapse
Affiliation(s)
- Sonia Shinhmar
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK; (S.S.); (J.S.); (K.L.J.); (P.B.)
| | - Judith Schaf
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK; (S.S.); (J.S.); (K.L.J.); (P.B.)
| | - Katie Lloyd Jones
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK; (S.S.); (J.S.); (K.L.J.); (P.B.)
| | - Olivier E. Pardo
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK;
| | - Philip Beesley
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK; (S.S.); (J.S.); (K.L.J.); (P.B.)
| | - Robin S. B. Williams
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK; (S.S.); (J.S.); (K.L.J.); (P.B.)
| |
Collapse
|
26
|
Liu W, Ma Y, He Y, Liu Y, Guo Z, He J, Han X, Hu Y, Li M, Jiang R, Wang S. Discovery of Novel p53-MDM2 Inhibitor (RG7388)-Conjugated Platinum IV Complexes as Potent Antitumor Agents. J Med Chem 2024; 67:9645-9661. [PMID: 38776419 DOI: 10.1021/acs.jmedchem.4c00784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
While a number of p53-MDM2 inhibitors have progressed into clinical trials for the treatment of cancer, their progression has been hampered by a variety of problems, including acquired drug resistance, dose-dependent toxicity, and limited clinical efficiency. To make more progress, we integrated the advantages of MDM2 inhibitors and platinum drugs to construct novel PtIV-RG7388 (a selective MDM2 inhibitor) complexes. Most complexes, especially 5a and 5b, displayed greatly improved antiproliferative activity against both wild-type and mutated p53 cancer cells. Remarkably, 5a exhibited potent in vivo tumor growth inhibition in the A549 xenograft model (66.5%) without apparent toxicity. It arrested the cell cycle at both the S phase and the G2/M phase and efficiently induced apoptosis via the synergistic effects of RG7388 and cisplatin. Altogether, PtIV-RG7388 complex 5a exhibited excellent in vitro and in vivo antitumor activities, highlighting the therapeutic potential of PtIV-RG7388 complexes as antitumor agents.
Collapse
Affiliation(s)
- Wei Liu
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
- Faculty of Pharmacy, School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Yi Ma
- Faculty of Pharmacy, School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Youyou He
- Faculty of Pharmacy, School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Yanhong Liu
- Faculty of Pharmacy, School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Zhongjie Guo
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jin He
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xiaodong Han
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yujiao Hu
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Muqiong Li
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ru Jiang
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Shengzheng Wang
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
27
|
Nguyen T, Aida T, Iijima‐Yamashita Y, Tamai M, Nagamachi A, Kagami K, Komatsu C, Kasai S, Akahane K, Goi K, Inaba T, Sanada M, Inukai T. Application of prime editing system to introduce TP53 R248Q hotspot mutation in acute lymphoblastic leukemia cell line. Cancer Sci 2024; 115:1924-1935. [PMID: 38549229 PMCID: PMC11145152 DOI: 10.1111/cas.16162] [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: 08/15/2023] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 06/04/2024] Open
Abstract
In childhood acute lymphoblastic leukemia (ALL), TP53 gene mutation is associated with chemoresistance in a certain population of relapsed cases. To directly verify the association of TP53 gene mutation with chemoresistance of relapsed childhood ALL cases and improve their prognosis, the development of appropriate human leukemia models having TP53 mutation in the intrinsic gene is required. Here, we sought to introduce R248Q hotspot mutation into the intrinsic TP53 gene in an ALL cell line, 697, by applying a prime editing (PE) system, which is a versatile genome editing technology. The PE2 system uses an artificial fusion of nickase Cas9 and reverse-transcriptase to directly place new genetic information into a target site through a reverse transcriptase template in the prime editing guide RNA (pegRNA). Moreover, in the advanced PE3b system, single guide RNA (sgRNA) matching the edited sequence is also introduced to improve editing efficiency. The initially obtained MDM2 inhibitor-resistant PE3b-transfected subline revealed disrupted p53 transactivation activity, reduced p53 target gene expression, and acquired resistance to chemotherapeutic agents and irradiation. Although the majority of the subline acquired the designed R248Q and adjacent silent mutations, the insertion of the palindromic sequence in the scaffold hairpin structure of pegRNA and the overlap of the original genomic DNA sequence were frequently observed. Targeted next-generation sequencing reconfirmed frequent edit errors in both PE2 and PE3b-transfected 697 cells, and it revealed frequent successful edits in HEK293T cells. These observations suggest a requirement for further modification of the PE2 and PE3b systems for accurate editing in leukemic cells.
Collapse
Affiliation(s)
- Thao Nguyen
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Tomomi Aida
- McGovern Institute for Brain ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Yuka Iijima‐Yamashita
- Department of Advanced DiagnosisClinical Research Center, NHO Nagoya Medical CenterNagoyaJapan
| | - Minori Tamai
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Akiko Nagamachi
- Department of Molecular Oncology and Leukemia Program ProjectResearch Institute for Radiation Biology and Medicine, Hiroshima UniversityHigashihiroshimaJapan
| | - Keiko Kagami
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Chiaki Komatsu
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Shin Kasai
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Koshi Akahane
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Kumiko Goi
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Toshiya Inaba
- Department of Molecular Oncology and Leukemia Program ProjectResearch Institute for Radiation Biology and Medicine, Hiroshima UniversityHigashihiroshimaJapan
| | - Masashi Sanada
- Department of Advanced DiagnosisClinical Research Center, NHO Nagoya Medical CenterNagoyaJapan
| | - Takeshi Inukai
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| |
Collapse
|
28
|
Fan YL, Jin JX, Zhu J, Ruan HB, Huang JQ. Extracellular vesicles of Bifidobacterium longum reverse the acquired carboplatin resistance in ovarian cancer cells via p53 phosphorylation on Ser15. Kaohsiung J Med Sci 2024; 40:530-541. [PMID: 38647095 DOI: 10.1002/kjm2.12837] [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: 01/24/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
We previously found that the relative abundance of Bifidobacterium was increased after chemotherapy; however, the role of Bifidobacterium longum in chemotherapeutic drug resistance in ovarian cancer (OVC) remains unclear. This study aimed to understand the potential effects and mechanism of B. longum extracellular vesicles (B. longum-EVs) on carboplatin (CBP) resistance in OVC. Eight normal and 11 ovarian tissues were collected and the expression of B. longum genomic DNA and its association with acquired CBP resistance in OVC patients was determined. After isolating EVs by ultracentrifugation from B. longum (ATCC 15707), CBP-resistant A2780 cells were treated with PBS, CBP, B. longum-EVs, or CBP + B. longum-EVs, and subsequently analyzed by CCK-8, Edu staining, Annexin V/PI double staining, wound healing, and Transwell assays to detect cell viability, proliferation, apoptosis, migration, and invasion, respectively. MRP1, ATP7A, ATP7B, and p53 expression as well as p53 phosphorylation were measured by western blot analysis. S15A mutation of p53 was assessed to examine the potential role of p53 Ser15 phosphorylation in CBP-resistant OVC. B. longum levels were elevated and positively associated with CBP resistance in OVC patients. Only high concentrations of B. longum-EVs attenuated A2780 cell proliferation, apoptosis, migration, and invasion. B. longum-EVs exposure significantly enhanced the sensitivity of CBP-resistant A2780 cells to CBP and decreased the expression of drug resistance-related proteins. The effect of B. longum-EVs on reversing CBP resistance was completely inhibited by S15A mutation of p53. B. longum-EVs enhanced the sensitivity of OVC cells to CBP through p53 phosphorylation on Ser15.
Collapse
Affiliation(s)
- Yun-Long Fan
- Departments of Gynaecology and Obstetrics, The First People's Hospital of Wenling, Wenling, China
| | - Jia-Xi Jin
- Departments of Gynaecology and Obstetrics, The First People's Hospital of Wenling, Wenling, China
| | - Jun Zhu
- Departments of Gynaecology and Obstetrics, The First People's Hospital of Wenling, Wenling, China
| | - Hai-Bo Ruan
- Departments of Gynaecology and Obstetrics, The First People's Hospital of Wenling, Wenling, China
| | - Jin-Qun Huang
- Departments of Gynaecology and Obstetrics, The First People's Hospital of Wenling, Wenling, China
| |
Collapse
|
29
|
Bianchi M, Paravani EV, Acosta MG, Odetti LM, Simoniello MF, Poletta GL. Pesticide-induced alterations in zebrafish (Danio rerio) behavior, histology, DNA damage and mRNA expression: An integrated approach. Comp Biochem Physiol C Toxicol Pharmacol 2024; 280:109895. [PMID: 38479676 DOI: 10.1016/j.cbpc.2024.109895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/20/2024] [Accepted: 03/09/2024] [Indexed: 03/22/2024]
Abstract
To assess the impact of glyphosate and 2,4-D herbicides, as well as the insecticide imidacloprid, both individually and in combination, the gills of adult zebrafish were used due to their intimate interaction with chemicals diluted in water. Bioassays were performed exposing the animals to the different pesticides and their mixture for 96 h. The behavior of the fish was analyzed, a histological examination of the gills was carried out, and the genotoxic effects were also analyzed by means of the comet assay (CA) and the change in the expression profiles of genes involved in the pathways of the oxidative stress and cellular apoptosis. The length traveled and the average speed of the control fish, compared to those exposed to the pesticides and mainly those exposed to the mixture, were significantly greater. All the groups exposed individually exhibited a decrease in thigmotaxis time, indicating a reduction in the behavior of protecting themselves from predators. Histological analysis revealed significant differences in the structures of the gill tissues. The quantification of the histological lesions showed mild lesions in the fish exposed to imidacloprid, moderate to severe lesions for glyphosate, and severe lesions in the case of 2,4-D and the mixture of pesticides. The CA revealed the sensitivity of gill cells to DNA damage following exposure to glyphosate, 2,4-D, imidacloprid and the mixture. Finally, both genes involved in the oxidative stress pathway and those related to the cell apoptosis pathway were overexpressed, while the ogg1 gene, involved in DNA repair, was downregulated.
Collapse
Affiliation(s)
- M Bianchi
- Laboratorio de Química Ambiental, Facultad de Ingeniería, Universidad Nacional de Entre Ríos, Oro Verde, Argentina.
| | - E V Paravani
- Laboratorio de Química Ambiental, Facultad de Ingeniería, Universidad Nacional de Entre Ríos, Oro Verde, Argentina
| | - M G Acosta
- Laboratorio de Química Ambiental, Facultad de Ingeniería, Universidad Nacional de Entre Ríos, Oro Verde, Argentina
| | - L M Odetti
- Cátedra de Toxicología, Farmacología y Bioquímica Legal, FBCB-UNL, Ciudad Universitaria, Santa Fe, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CABA, Argentina
| | - M F Simoniello
- Cátedra de Toxicología, Farmacología y Bioquímica Legal, FBCB-UNL, Ciudad Universitaria, Santa Fe, Argentina
| | - G L Poletta
- Cátedra de Toxicología, Farmacología y Bioquímica Legal, FBCB-UNL, Ciudad Universitaria, Santa Fe, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CABA, Argentina
| |
Collapse
|
30
|
Perrone G, Rigacci L, Roviello G, Landini I, Fabbri A, Iovino L, Puccini B, Cencini E, Orciuolo E, Bocchia M, Bosi A, Mini E, Nobili S. Validation of single nucleotide polymorphisms potentially related to R-CHOP resistance in diffuse large B-cell lymphoma patients. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:21. [PMID: 38835350 PMCID: PMC11149109 DOI: 10.20517/cdr.2024.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/30/2024] [Accepted: 05/16/2024] [Indexed: 06/06/2024]
Abstract
Aim: Diffuse large B-cell lymphoma (DLBCL) is the most common B-cell non-Hodgkin lymphoma (NHL). Despite the availability of clinical and molecular algorithms applied for the prediction of prognosis, in up to 30%-40% of patients, intrinsic or acquired drug resistance occurs. Constitutional genetics may help to predict R-CHOP resistance. This study aimed to validate previously identified single nucleotide polymorphisms (SNPs) in the literature as potential predictors of R-CHOP resistance in DLBCL patients, SNPs. Methods: Twenty SNPs, involved in R-CHOP pharmacokinetics/pharmacodynamics or other pathobiological processes, were investigated in 185 stage I-IV DLBCL patients included in a multi-institution pharmacogenetic study to validate their previously identified correlations with resistance to R-CHOP. Results: Correlations between rs2010963 (VEGFA gene) and sex (P = 0.046), and rs1625895 (TP53 gene) and stage (P = 0.003) were shown. After multivariate analyses, a concordant effect (i.e., increased risk of disease progression and death) was observed for rs1883112 (NCF4 gene) and rs1800871 (IL10 gene). When patients were grouped according to the revised International Prognostic Index (R-IPI), both these SNPs further discriminated progression-free survival (PFS) and overall survival (OS) of the R-IPI-1-2 subgroup. Overall, patients harboring the rare allele showed shorter PFS and OS compared with wild-type patients. Conclusions: Two out of the 20 study SNPs were validated. Thus, these results support the role of previously identified rs1883112 and rs1800871 in predicting DLBCL resistance to R-CHOP and highlight their ability to further discriminate the prognosis of R-IPI-1-2 patients. These data point to the need to also focus on host genetics for a more comprehensive assessment of DLBCL patient outcomes in future prospective trials.
Collapse
Affiliation(s)
- Gabriele Perrone
- Department of Health Sciences, University of Florence, Florence 50139, Italy
| | - Luigi Rigacci
- Research Unit of Hematology, Department of Medicine and Surgery, Campus Biomedico University, Rome 00128, Italy
| | | | - Ida Landini
- Department of Health Sciences, University of Florence, Florence 50139, Italy
| | - Alberto Fabbri
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena 53100, Italy
| | - Lorenzo Iovino
- Unit of Hematology, Santa Chiara University Hospital, University of Pisa, Pisa 56126, Italy
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109-4433, USA
| | - Benedetta Puccini
- Unit of Hematology, Careggi University-Hospital, Florence 50134, Italy
| | - Emanuele Cencini
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena 53100, Italy
| | - Enrico Orciuolo
- Unit of Hematology, Santa Chiara University Hospital, University of Pisa, Pisa 56126, Italy
| | - Monica Bocchia
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena 53100, Italy
| | - Alberto Bosi
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Enrico Mini
- Department of Health Sciences, University of Florence, Florence 50139, Italy
- Authors contributed equally
| | - Stefania Nobili
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence 50139, Italy
- Authors contributed equally
| |
Collapse
|
31
|
Abdel-Megeed RM, Abdel-Hamid AHZ, Kadry MO. Titanium dioxide nanostructure-loaded Adriamycin surmounts resistance in breast cancer therapy: ABCA/P53/C-myc crosstalk. Future Sci OA 2024; 10:FSO979. [PMID: 38827789 PMCID: PMC11140649 DOI: 10.2144/fsoa-2023-0107] [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] [Indexed: 06/05/2024] Open
Abstract
Aim: To clarify the alternation of gene expression responsible for resistance of Adriamycin (ADR) in rats, in addition to investigation of a novel promising drug-delivery system using titanium dioxide nanoparticles loaded with ADR (TiO2-ADR). Method: Breast cancer was induced in female Sprague-Dawley rats, followed by treatment with ADR (5 mg/kg) or TiO2-ADR (2 mg/kg) for 1 month. Results: Significant improvements in both zinc and calcium levels were observed with TiO2-ADR treatment. Gene expression of ATP-binding cassette transporter membrane proteins (ABCA1 & ABCG1), P53 and Jak-2 showed a significant reduction and overexpression of the C-myc in breast cancer-induced rats. TiO2-ADR demonstrated a notable ability to upregulate these genes. Conclusion: TiO2-ADR could be a promising drug-delivery system for breast cancer therapy.
Collapse
Affiliation(s)
- Rehab M Abdel-Megeed
- Therapeutic Chemistry Department, Pharmaceutical & Drug Industries Research Institute, National Research Center, El Buhouth St., Dokki, Cairo, 12622, Egypt
| | - Abdel-Hamid Z Abdel-Hamid
- Therapeutic Chemistry Department, Pharmaceutical & Drug Industries Research Institute, National Research Center, El Buhouth St., Dokki, Cairo, 12622, Egypt
| | - Mai O Kadry
- Therapeutic Chemistry Department, Pharmaceutical & Drug Industries Research Institute, National Research Center, El Buhouth St., Dokki, Cairo, 12622, Egypt
| |
Collapse
|
32
|
Nova P, Gomes AM, Costa-Pinto AR. It comes from the sea: macroalgae-derived bioactive compounds with anti-cancer potential. Crit Rev Biotechnol 2024; 44:462-476. [PMID: 36842998 DOI: 10.1080/07388551.2023.2174068] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 01/14/2023] [Indexed: 02/28/2023]
Abstract
Nature derived compounds represent a valuable source of bioactive molecules with enormous potential. The sea is one of the richest environments, full of skilled organisms, where algae stand out due to their unique characteristics. Marine macroalgae adapt their phenotypic characteristics, such as chemical composition, depending on the environmental conditions where they live. The compounds produced by these organisms show tremendous potential to be used in the biomedical field, due to their antioxidant, anti-inflammatory, immunomodulatory, and anti-cancer properties.Cancer is one of the deadliest diseases in the world, and the lack of effective treatments highlights the urgent need for the development of new therapeutic strategies. This review provides an overview of the current advances regarding the anti-cancer activity of the three major groups of marine macroalgae, i.e., red algae (Rhodophyta), brown algae (Phaeophyceae), and green algae (Chlorophyta) on pancreatic, lung, breast, cervical, colorectal, liver, and gastric cancers as well as leukemia and melanoma. In addition, future perspectives, and limitations regarding this field of work are also discussed.
Collapse
Affiliation(s)
- Paulo Nova
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ana Maria Gomes
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Ana R Costa-Pinto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Instituto de Patologia Molecular e Imunologia da Universidade do Porto, Porto, Portugal
| |
Collapse
|
33
|
Chauhan S, Jaiswal S, Jakhmola V, Singh B, Bhattacharya S, Garg M, Sengupta S. Potential role of p53 deregulation in modulating immune responses in human malignancies: A paradigm to develop immunotherapy. Cancer Lett 2024; 588:216766. [PMID: 38408603 PMCID: PMC7615729 DOI: 10.1016/j.canlet.2024.216766] [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: 01/19/2024] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
The crucial role played by the oncogenic expression of TP53, stemming from mutation or amyloid formation, in various human malignancies has been extensively studied over the past two decades. Interestingly, the potential role of TP53 as a crucial player in modulating immune responses has provided new insight into the field of cancer biology. The loss of p53's transcriptional functions and/or the acquisition of tumorigenic properties can efficiently modulate the recruitment and functions of myeloid and lymphoid cells, ultimately leading to the evasion of immune responses in human tumors. Consequently, the oncogenic nature of the tumor suppressor p53 can dynamically alter the function of immune cells, providing support for tumor progression and metastasis. This review comprehensively explores the dual role of p53 as both the guardian of the genome and an oncogenic driver, especially in the context of regulation of autophagy, apoptosis, the tumor microenvironment, immune cells, innate immunity, and adaptive immune responses. Additionally, the focus of this review centers on how p53 status in the immune response can be harnessed for the development of tailored therapeutic strategies and their potential application in immunotherapy against human malignancies.
Collapse
Affiliation(s)
- Shivi Chauhan
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noda, 201313, India
| | - Shivani Jaiswal
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noda, 201313, India
| | - Vibhuti Jakhmola
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noda, 201313, India
| | - Bhavana Singh
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noda, 201313, India
| | - Sujata Bhattacharya
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noda, 201313, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noda, 201313, India.
| | - Shinjinee Sengupta
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noda, 201313, India.
| |
Collapse
|
34
|
Podolski-Renić A, Čipak Gašparović A, Valente A, López Ó, Bormio Nunes JH, Kowol CR, Heffeter P, Filipović NR. Schiff bases and their metal complexes to target and overcome (multidrug) resistance in cancer. Eur J Med Chem 2024; 270:116363. [PMID: 38593587 DOI: 10.1016/j.ejmech.2024.116363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/15/2024] [Accepted: 03/25/2024] [Indexed: 04/11/2024]
Abstract
Overcoming multidrug resistance (MDR) is one of the major challenges in cancer therapy. In this respect, Schiff base-related compounds (bearing a R1R2CNR3 bond) gained high interest during the past decades. Schiff bases are considered privileged ligands for various reasons, including the easiness of their preparation and the possibility to form complexes with almost all transition metal ions. Schiff bases and their metal complexes exhibit many types of biological activities and are used for the treatment and diagnosis of various diseases. Until now, 13 Schiff bases have been investigated in clinical trials for cancer treatment and hypoxia imaging. This review represents the first collection of Schiff bases and their complexes which demonstrated MDR-reversal activity. The areas of drug resistance covered in this article involve: 1) Modulation of ABC transporter function, 2) Targeting lysosomal ABCB1 overexpression, 3) Circumvention of ABC transporter-mediated drug efflux by alternative routes of drug uptake, 4) Selective activity against MDR cancer models (collateral sensitivity), 5) Targeting GSH-detoxifying systems, 6) Overcoming apoptosis resistance by inducing necrosis and paraptosis, 7) Reactivation of mutated p53, 8) Restoration of sensitivity to DNA-damaging anticancer therapy, and 9) Overcoming drug resistance through modulation of the immune system. Through this approach, we would like to draw attention to Schiff bases and their metal complexes representing highly interesting anticancer drug candidates with the ability to overcome MDR.
Collapse
Affiliation(s)
- Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Serbia
| | | | - Andreia Valente
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa, Portugal
| | - Óscar López
- Departamento de Química Organica, Facultad de Química, Universidad de Sevilla, Sevilla, Spain
| | - Julia H Bormio Nunes
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria; Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christian R Kowol
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Petra Heffeter
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| | - Nenad R Filipović
- Department of Chemistry and Biochemistry, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
35
|
Hosseini SA, Mirzaei SA, Kermani S, Yaghoobi H. Valproate modulates the activity of multidrug resistance efflux pumps, as a chemoresistance factor in gastric cancer cells. Mol Biol Rep 2024; 51:427. [PMID: 38498238 DOI: 10.1007/s11033-024-09284-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/24/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Drug resistance is one of the most critical problems in gastric cancer therapy. This study was performed to investigate the valproic acid effects on the proliferation of sensitive and resistant cell lines of human gastric cancer, and to explore the mechanism of the agent on multi drug resistance and apoptosis genes. METHODS The cytotoxicity effect of valproic acid on the EPG85.257 and EPG85.257RDB cells was assessed by the MTT assay, and the IC50 concentration was evaluated. Apoptosis, genotoxicity, and drug resistance pump activity were evaluated using comet assay, Real-time PCR, and flow cytometry, respectively. Cell proliferation was assayed using a scratch test. RESULTS Dose-dependent toxicity was recorded after treatment of cells with valproic acid. Valproic acid represented a significant growth inhibition on EPG85.257 cells with IC50 values of 5.84 µM and 4.78 µM after 48 h and 72 h treatment, respectively. In contrast, the drug-resistant counterpart represented 8.7 µM and 7.02 µM IC50 values after the same treatment time. Valproic acid induced PTEN, Bcl2, P53, Bax, P21, and caspase3 expression in EPG85.257 cells, whereas p21, p53, PTEN, and ABCB1 were overexpressed in EPG5.257RDB. Valproic acid hindered cell migration in both cell lines (P < 0.01). Valproate genotoxicity was significantly higher in the parent cells than in their resistant EPG85.257RDB counterparts. Valproate led to a 62% reduction in the daunorubicin efflux of the MDR1 pump activity. CONCLUSIONS Valproate can affect drug resistance in gastric cancer via a unique mechanism independent of MDR1 expression.
Collapse
Affiliation(s)
- Sayedeh Azimeh Hosseini
- Student Research Commitee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Department of Medical Biotechnology, School of Advanced Technology, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyed Abbas Mirzaei
- Department of Medical Biotechnology, School of Advanced Technology, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Shahriar Kermani
- Student Research Commitee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hajar Yaghoobi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
36
|
Adico MD, Bayala B, Zoure AA, Lagarde A, Bazie JT, Traore L, Buñay J, Yonli AT, Djigma F, Bambara HA, Baron S, Simporé J, Lobaccaro JMA. In vitro activities and mechanisms of action of anti-cancer molecules from African medicinal plants: a systematic review. Am J Cancer Res 2024; 14:1376-1401. [PMID: 38590420 PMCID: PMC10998760 DOI: 10.62347/auhb5811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/27/2023] [Indexed: 04/10/2024] Open
Abstract
Cancer is one of the leading causes of death worldwide. In recent years, African countries have been faced with a rapid increase in morbidity and mortality due to this pathology. Management is often complicated by the high treatment costs, side effects and the increasing occurrence of resistance to treatments. The identification of new active ingredients extracted from endemic medicinal plants is definitively an interesting approach for the implementation of new therapeutic strategies: their extraction is often lower cost; their identification is based on an ethnobotanical history and a tradipratic approach; their use by low-income populations is simpler; this can help in the development of new synthetic molecules that are more active, more effective and with fewer side effects. The objective of this review is to document the molecules derived from African medicinal plants whose in vitro anti-cancer activities and the mechanisms of molecular actions have been identified. From the scientific databases Science Direct, PubMed and Google Scholar, we searched for publications on compounds isolated from African medicinal plants and having activity on cancer cells in culture. The data were analyzed in particular with regard to the cytotoxicity of the compounds and their mode of action. A total of 90 compounds of these African medicinal plants were selected. They come from nine chemical groups: alkaloids, flavonoids, polyphenols, quinones, saponins, steroids, terpenoids, xanthones and organic sulfides. These compounds have been associated with several cellular effects: i) Cytotoxicity, including caspase activation, alteration of mitochondrial membrane potential, and/or induction of reactive oxygen species (ROS); ii) Anti-angiogenesis; iii) Anti-metastatic properties. This review points out that the cited African plants are rich in active ingredients with anticancer properties. It also stresses that screening of these anti-tumor active ingredients should be continued at the continental scale. Altogether, this work provides a rational basis for the selection of phytochemical compounds for use in clinical trials.
Collapse
Affiliation(s)
- Marc Dw Adico
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
| | - Bagora Bayala
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
- Ecole Normale Supérieure Koudougou, Burkina Faso
| | - Abdou A Zoure
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
- Laboratoire de recherches Biomédicales (LaReBio), Département de santé publique et biomédicale, Institut de Recherche en Sciences de la Santé (IRSS/CNRST) Ouagadougou, Burkina Faso
| | - Aurélie Lagarde
- Institute Génétique, Reproduction, Développement, UMR CNRS 6293, INSERM U1103, Université Clermont Auvergne 28, Place Henri Dunant, BP38, F63001, Clermont-Ferrand, France
| | - Jean Tv Bazie
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
- Département des Substances Naturelles (DSN), Institut de Recherche en Sciences et Technologies Appliquées (IRSAT) Ouagadougou, Burkina Faso
| | - Lassina Traore
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
| | - Julio Buñay
- Institute Génétique, Reproduction, Développement, UMR CNRS 6293, INSERM U1103, Université Clermont Auvergne 28, Place Henri Dunant, BP38, F63001, Clermont-Ferrand, France
| | - Albert T Yonli
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
| | - Florencia Djigma
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
| | - Hierrhum A Bambara
- Service d'oncologie, Centre hospitalier universitaire BOGODOGO, Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
| | - Silvère Baron
- Institute Génétique, Reproduction, Développement, UMR CNRS 6293, INSERM U1103, Université Clermont Auvergne 28, Place Henri Dunant, BP38, F63001, Clermont-Ferrand, France
| | - Jacques Simporé
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
- Faculté de médecine, Université Saint Thomas d'Aquin (USTA) Ouagadougou, Burkina Faso
| | - Jean-Marc A Lobaccaro
- Institute Génétique, Reproduction, Développement, UMR CNRS 6293, INSERM U1103, Université Clermont Auvergne 28, Place Henri Dunant, BP38, F63001, Clermont-Ferrand, France
| |
Collapse
|
37
|
Benedetti F, Mongodin EF, Badger JH, Munawwar A, Cellini A, Yuan W, Silvestri G, Kraus CN, Marini S, Rathinam CV, Salemi M, Tettelin H, Gallo RC, Zella D. Bacterial DnaK reduces the activity of anti-cancer drugs cisplatin and 5FU. J Transl Med 2024; 22:269. [PMID: 38475767 PMCID: PMC10935962 DOI: 10.1186/s12967-024-05078-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 03/07/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Chemotherapy is a primary treatment for cancer, but its efficacy is often limited by cancer-associated bacteria (CAB) that impair tumor suppressor functions. Our previous research found that Mycoplasma fermentans DnaK, a chaperone protein, impairs p53 activities, which are essential for most anti-cancer chemotherapeutic responses. METHODS To investigate the role of DnaK in chemotherapy, we treated cancer cell lines with M. fermentans DnaK and then with commonly used p53-dependent anti-cancer drugs (cisplatin and 5FU). We evaluated the cells' survival in the presence or absence of a DnaK-binding peptide (ARV-1502). We also validated our findings using primary tumor cells from a novel DnaK knock-in mouse model. To provide a broader context for the clinical significance of these findings, we investigated human primary cancer sequencing datasets from The Cancer Genome Atlas (TCGA). We identified F. nucleatum as a CAB carrying DnaK with an amino acid composition highly similar to M. fermentans DnaK. Therefore, we investigated the effect of F. nucleatum DnaK on the anti-cancer activity of cisplatin and 5FU. RESULTS Our results show that both M. fermentans and F. nucleatum DnaKs reduce the effectiveness of cisplatin and 5FU. However, the use of ARV-1502 effectively restored the drugs' anti-cancer efficacy. CONCLUSIONS Our findings offer a practical framework for designing and implementing novel personalized anti-cancer strategies by targeting specific bacterial DnaKs in patients with poor response to chemotherapy, underscoring the potential for microbiome-based personalized cancer therapies.
Collapse
Affiliation(s)
- Francesca Benedetti
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Emmanuel F Mongodin
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jonathan H Badger
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Arshi Munawwar
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ashley Cellini
- Pathology Biorepository Shared Service, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA
| | - Weirong Yuan
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Giovannino Silvestri
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Simone Marini
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
- Department of Epidemiology, University of Florida, Gainesville, FL, USA
| | - Chozha V Rathinam
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marco Salemi
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
- Department of Pathology, University of Florida, Gainesville, FL, USA
| | - Hervé Tettelin
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Robert C Gallo
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Davide Zella
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
38
|
Basha NJ, Mohan RM. Insight on Heterocycles as p53‐MDM2 Protein‐Protein Interaction Inhibitors: Molecular Mechanism for p53 Activation. ChemistrySelect 2024; 9. [DOI: 10.1002/slct.202304525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/20/2024] [Indexed: 01/03/2025]
Abstract
AbstractTranscription factor p53, also known as tumor suppressor protein. Encoded by the TP53 gene, the guardian of genome p53 regulates many gene pathways. Nevertheless, the molecular mechanisms of p53 functioning have been known for a few decades, and the exact role of p53 in cancer therapy is unclear. Also, comprehensive literature on heterocycles as p53‐MDM2 protein‐protein interaction inhibitors is limited. This review covers the molecular mechanism for the p53‐MDM2 interaction and its inhibition by the heterocyclic small molecules. We hope the present comprehensive study will help to develop heterocycles as anticancer drugs that induce apoptosis in tumor cells.
Collapse
Affiliation(s)
- N. Jeelan Basha
- Department of Chemistry Indian Academy Degree College-Autonomous Bengaluru India- 560043
| | - R. M. Mohan
- Department of Chemistry Indian Academy Degree College-Autonomous Bengaluru India- 560043
| |
Collapse
|
39
|
Smoots SG, Schreiber AR, Jackson MM, Bagby SM, Dominguez ATA, Dus ED, Binns CA, MacBeth M, Whitty PA, Diamond JR, Pitts TM. Overcoming doxorubicin resistance in triple-negative breast cancer using the class I-targeting HDAC inhibitor bocodepsin/OKI-179 to promote apoptosis. Breast Cancer Res 2024; 26:35. [PMID: 38429789 PMCID: PMC10908182 DOI: 10.1186/s13058-024-01799-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with a poor prognosis. Doxorubicin is part of standard curative therapy for TNBC, but chemotherapy resistance remains an important clinical challenge. Bocodepsin (OKI-179) is a small molecule class I histone deacetylase (HDAC) inhibitor that promotes apoptosis in TNBC preclinical models. The purpose of this study was to investigate the combination of bocodepsin and doxorubicin in preclinical TNBC models and evaluate the impact on terminal cell fate, including apoptosis and senescence. METHODS TNBC cell lines were treated with doxorubicin and CellTiter-Glo was used to assess proliferation and determine doxorubicin sensitivity. Select cell lines were treated with OKI-005 (in vitro version of bocodepsin) and doxorubicin and assessed for proliferation, apoptosis as measured by Annexin V/PI, and cell cycle by flow cytometry. Immunoblotting was used to assess changes in mediators of apoptosis, cell cycle arrest, and senescence. Senescence was measured by the senescence-associated β-galactosidase assay. An MDA-MB-231 xenograft in vivo model was treated with bocodepsin, doxorubicin, or the combination and assessed for inhibition of tumor growth. shRNA knockdown of p53 was performed in the CAL-51 cell line and proliferation, apoptosis and senescence were assessed in response to combination treatment. RESULTS OKI-005 and doxorubicin resulted in synergistic antiproliferative activity in TNBC cells lines regardless of p53 mutation status. The combination led to increased apoptosis and decreased senescence. In vivo, the combination resulted in increased tumor growth inhibition compared to either single agent. shRNA knock-down of p53 led to increased doxorubicin-induced senescence that was decreased with the addition of OKI-005 in vitro. CONCLUSION The addition of bocodepsin to doxorubicin resulted in synergistic antiproliferative activity in vitro, improved tumor growth inhibition in vivo, and promotion of apoptosis which makes this a promising combination to overcome doxorubicin resistance in TNBC. Bocodepsin is currently in clinical development and has a favorable toxicity profile compared to other HDAC inhibitors supporting the feasibility of evaluating this combination in patients with TNBC.
Collapse
Affiliation(s)
- Stephen G Smoots
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Anna R Schreiber
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Marilyn M Jackson
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Stacey M Bagby
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Adrian T A Dominguez
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Evan D Dus
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Cameron A Binns
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Morgan MacBeth
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Phaedra A Whitty
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Jennifer R Diamond
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Todd M Pitts
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA.
| |
Collapse
|
40
|
Naeimzadeh Y, Tajbakhsh A, Fallahi J. Understanding the prion-like behavior of mutant p53 proteins in triple-negative breast cancer pathogenesis: The current therapeutic strategies and future directions. Heliyon 2024; 10:e26260. [PMID: 38390040 PMCID: PMC10881377 DOI: 10.1016/j.heliyon.2024.e26260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/20/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024] Open
Abstract
Breast cancer (BC) is viewed as a significant public health issue and is the primary cause of cancer-related deaths among women worldwide. Triple-negative breast cancer (TNBC) is a particularly aggressive subtype that predominantly affects young premenopausal women. The tumor suppressor p53 playsa vital role in the cellular response to DNA damage, and its loss or mutations are commonly present in many cancers, including BC. Recent evidence suggests that mutant p53 proteins can aggregate and form prion-like structures, which may contribute to the pathogenesis of different types of malignancies, such as BC. This review provides an overview of BC molecular subtypes, the epidemiology of TNBC, and the role of p53 in BC development. We also discuss the potential implications of prion-like aggregation in BC and highlight future research directions. Moreover, a comprehensive analysis of the current therapeutic approaches targeting p53 aggregates in BC treatment is presented. Strategies including small molecules, chaperone inhibitors, immunotherapy, CRISPR-Cas9, and siRNA are discussed, along with their potential benefits and drawbacks. The use of these approaches to inhibit p53 aggregation and degradation represents a promising target for cancer therapy. Future investigations into the efficacy of these approaches against various p53 mutations or binding to non-p53 proteins should be conducted to develop more effective and personalized therapies for BC treatment.
Collapse
Affiliation(s)
- Yasaman Naeimzadeh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
| |
Collapse
|
41
|
Garg A, Kumar G, Singh V, Sinha S. Doxorubicin catalyses self-assembly of p53 by phase separation. Curr Res Struct Biol 2024; 7:100133. [PMID: 38435052 PMCID: PMC10906149 DOI: 10.1016/j.crstbi.2024.100133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/29/2024] [Accepted: 02/15/2024] [Indexed: 03/05/2024] Open
Abstract
Liquid-liquid phase separation plays a crucial role in cellular physiology, as it leads to the formation of membrane-less organelles in response to various internal stimuli, contributing to various cellular functions. However, the influence of exogenous stimuli on this process in the context of disease intervention remains unexplored. In this current investigation, we explore the impact of doxorubicin on the abnormal self-assembly of p53 using a combination of biophysical and imaging techniques. Additionally, we shed light on the potential mechanisms behind chemoresistance in cancer cells carrying mutant p53. Our findings reveal that doxorubicin co-localizes with both wild-type p53 (WTp53) and its mutant variants. Our in vitro experiments indicate that doxorubicin interacts with the N-terminal-deleted form of WTp53 (WTp53ΔNterm), inducing liquid-liquid phase separation, ultimately leading to protein aggregation. Notably, the p53 variants at the R273 position exhibit a propensity for phase separation even in the absence of doxorubicin, highlighting the destabilizing effects of point mutations at this position. The strong interaction between doxorubicin and p53 variants, along with its localization within the protein condensates, provides a potential explanation for the development of chemotherapy resistance. Collectively, our cellular and in vitro studies emphasize the role of exogenous agents in driving phase separation-mediated p53 aggregation.
Collapse
Affiliation(s)
- Ankush Garg
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector- 81, Mohali (SAS Nagar), Punjab, 140306, India
| | - Gaurav Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector- 81, Mohali (SAS Nagar), Punjab, 140306, India
| | - Varinder Singh
- Indian Institute of Science Education and Research, Sector- 81, Mohali (SAS Nagar), Punjab, 140306, India
| | - Sharmistha Sinha
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector- 81, Mohali (SAS Nagar), Punjab, 140306, India
| |
Collapse
|
42
|
Nawas MT, Kosuri S. Utility or futility? A contemporary approach to allogeneic hematopoietic cell transplantation for TP53-mutated MDS/AML. Blood Adv 2024; 8:553-561. [PMID: 38096805 PMCID: PMC10835231 DOI: 10.1182/bloodadvances.2023010417] [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: 09/26/2023] [Accepted: 12/07/2023] [Indexed: 01/25/2024] Open
Abstract
ABSTRACT TP 53-mutated myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) are among the most lethal malignancies, characterized by dismal outcomes with currently available therapies. Allogeneic hematopoietic cell transplantation (allo-HCT) is widely thought to be the only treatment option to offer durable disease control. However, outcomes with allo-HCT in this context are quite poor, calling into question the utility of transplantation. In this review, we summarize the latest data on allo-HCT outcomes in this subgroup, evaluating the limitations of available evidence; we review the molecular heterogeneity of this disease, delineating outcomes based on distinct biological features to aid in patient selection; and we critically examine whether allo-HCT should be routinely applied in this disease on the basis of currently available data. We propose that the exceptionally poor outcomes of patients with TP53-mutated MDS/AML with biallelic loss and/or adverse-risk cytogenetics should motivate randomized-controlled trials of HCT vs non-HCT to determine whether transplantation can prolong survival and/or positively impact other clinically relevant outcomes such as patient-reported outcomes or healthcare resource utilization in this disease subset. Without dedicated prospective randomized trials, selecting who may actually derive benefit from allo-HCT for TP53-mutated MDS/AML can be described as ambiguous guesswork and must be carefully contemplated.
Collapse
Affiliation(s)
- Mariam T. Nawas
- Hematopoietic Cellular Therapy Program, Department of Medicine, The University of Chicago Medicine, Chicago, IL
| | - Satyajit Kosuri
- Hematopoietic Cellular Therapy Program, Department of Medicine, The University of Chicago Medicine, Chicago, IL
| |
Collapse
|
43
|
Ben Rejeb S, Kouki N, Elfekih S, Cherif I, Khouni H. Prognostic significance of tumor suppressor protein p53 in prostate cancer. LA TUNISIE MEDICALE 2024; 102:111-115. [PMID: 38567478 PMCID: PMC11358809 DOI: 10.62438/tunismed.v102i2.4730] [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: 11/27/2023] [Accepted: 01/29/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION The p53 gene mutation is one of the most common genetic alterations in many cancers. In prostate cancer (PCa), it has been associated with a poor prognosis, tumor progression and aggressiveness. P53 mutation induces an abnormal protein expression in related tissues. AIM This study aimed to assess p53 expression using immunohistochemistry in PCa and to discuss its prognostic value. METHODS We have retrospectively collected all cases of PCa diagnosed in our pathology department between 2012 and 2022. An automatized immunohistochemical analysis was performed using monoclonal p53 antibody. For each case, we assessed the proportion of positive cells and the intensity of staining. P53 expression was considered abnormal when it was totally negative or overexpressed (>=50% of positive cells). RESULTS Twenty-four cases have been selected. Abnormal p53 expression was found in 42% of cases (P53 was overexpressed in 6cases and totally negative in 4 cases). Mean age of patients with p53 abnormal expression was 70years old. Patients with p53 abnormal expression had Gleason score >7 in 5 cases, ISUP grade >2 in 3 cases, peri-neural invasion in 8cases, capsule invasion in 9cases. All patients with p53 overexpression developed androgen resistance (p<0.01). CONCLUSION An aberrant expression profile of the p53 protein was observed in 42% of cases, and a statistically significant association was found with androgen resistance. Our results suggest a potential prognostic role of p53 in PCa.
Collapse
Affiliation(s)
- Sarra Ben Rejeb
- Department of pathology. Internal security forces hospital, Tunis, Tunisia
| | - Nadia Kouki
- Department of pathology. Internal security forces hospital, Tunis, Tunisia
| | - Sirine Elfekih
- Department of pathology. Internal security forces hospital, Tunis, Tunisia
| | - Ines Cherif
- Department of pathology. Internal security forces hospital, Tunis, Tunisia
| | - Hassen Khouni
- Department of urology. Internal security forces hospital, Tunis, Tunisia
| |
Collapse
|
44
|
Huang Y, Jiao Z, Fu Y, Hou Y, Sun J, Hu F, Yu S, Gong K, Liu Y, Zhao G. An overview of the functions of p53 and drugs acting either on wild- or mutant-type p53. Eur J Med Chem 2024; 265:116121. [PMID: 38194777 DOI: 10.1016/j.ejmech.2024.116121] [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: 11/02/2023] [Revised: 12/22/2023] [Accepted: 01/01/2024] [Indexed: 01/11/2024]
Abstract
TP53, also known as the "guardian of the genome," is an important tumor suppressor gene. It is encoded by the human genome and is associated with the development of diverse cancers. The p53 protein, encoded by TP53, functions in the cell to monitor DNA damage and prompts the cell to respond appropriately. When DNA is damaged, p53 halts the cell cycle, allowing cells to enter the repair state. If the repair is ineffective, p53 induces cell death via apoptosis. This prevents DNA damage transmission during cell division and reduces cancer risk. However, the p53 gene mutation compromises its function. This leads to the inability of cells to respond properly to DNA damage, which may result in cancer development. Mutations in p53 are widespread in diverse cancers, especially highly prevalent cancers, including breast, colon, and lung cancers. Despite the association between p53 mutations and cancer, researchers have discovered drugs and treatments that may reactivate mutated p53 function. Therefore, p53 remains an important area of research in cancer treatment and holds promise as a new direction for cancer therapy. In summary, TP53 is a vital tumor suppressor gene responsible for monitoring DNA damage and prompting cells to respond appropriately. This article summarizes drugs related to p53 and diverse strategies for discovering drugs that act on either wide or mutant p53. Herein, p53 is categorized into two types: wild and mutant type. Drugs are also classified according to diverse treatment strategies, enabling readers to differentiate between the two types of p53 and aiding in selecting the appropriate research direction. Additionally, this review offers a valuable reference for drug design.
Collapse
Affiliation(s)
- Yongmi Huang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, PR China.
| | - Zhihao Jiao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, PR China.
| | - Yuqing Fu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, PR China
| | - Yue Hou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, PR China
| | - Jinxiao Sun
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, PR China
| | - Feiran Hu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, PR China
| | - Shangzhe Yu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, PR China
| | - Kexin Gong
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, PR China
| | - Yiru Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, PR China
| | - Guisen Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, PR China.
| |
Collapse
|
45
|
Bhuia MS, Chowdhury R, Sonia FA, Biswas S, Ferdous J, El-Nashar HAS, El-Shazly M, Islam MT. Efficacy of Rotundic Acid and Its Derivatives as Promising Natural Anticancer Triterpenoids: A Literature-Based Study. Chem Biodivers 2024; 21:e202301492. [PMID: 38150556 DOI: 10.1002/cbdv.202301492] [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: 09/23/2023] [Revised: 12/21/2023] [Accepted: 12/25/2023] [Indexed: 12/29/2023]
Abstract
Rotundic acid (RA) is a naturally occurring pentacyclic triterpene with a multitude of pharmacological activities. The primary emphasis of this study is on summarizing the anticancer properties with the underlying mechanisms of RA and its derivatives, as well as the pharmacokinetic features. Data was collected (up to date as of November 10, 2023) from various reliable and authentic literatures by searching in different academic search engines, including PubMed, Springer Link, Scopus, Wiley Online, Web of Science, ScienceDirect, and Google Scholar. The findings imply that RA and its synthetic derivatives possess promising anti-cancer properties against breast, colorectal, liver, and cervical cancers in various preclinical pharmacological test systems. The results also indicate that RA and its derivatives demonstrated anticancer effects via a number of cellular mechanisms, including apoptotic cell death, inhibition of oxidative stress, anti-inflammatory effect, cytotoxicity, cell cycle arrest, anti-proliferative effect, anti-angiogenic effect, and inhibition of cancer cell migration and invasion. It has been proposed that RA and its derived compounds have the capability to serve as a hopeful chemotherapeutic agent, so further extensive clinical research is necessary.
Collapse
Affiliation(s)
- Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Fatema Akter Sonia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Shrabonti Biswas
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Jannatul Ferdous
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Heba A S El-Nashar
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, 11566, Abbassia, Cairo, Egypt
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, 11566, Abbassia, Cairo, Egypt
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| |
Collapse
|
46
|
Wu SY, Yu WJ, Chien TY, Ren YA, Chen CS, Chiang CS. Microglia-mediated drug substance transfer promotes chemoresistance in brain tumors: insights from an in vitro co-culture model using GCV/Tk prodrug system. Cancer Cell Int 2024; 24:35. [PMID: 38238749 PMCID: PMC10795391 DOI: 10.1186/s12935-024-03213-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 01/03/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND It is well known that tumor-associated macrophages (TAMs) play essential roles in brain tumor resistance to chemotherapy. However, the detailed mechanisms of how TAMs are involved in brain tumor resistance are still unclear and lack a suitable analysis model. METHODS A BV2 microglial cells with ALTS1C1 astrocytoma cells in vitro co-culture system was used to mimic the microglia dominating tumor stroma in the tumor invasion microenvironment and explore the interaction between microglia and brain tumor cells. RESULTS Our result suggested that microglia could form colonies with glioma cells under high-density culturing conditions and protect glioma cells from apoptosis induced by chemotherapeutic drugs. Moreover, this study demonstrates that microglia could hijack drug substances from the glioma cells and reduce the drug intensity of ALTS1C1 via direct contact. Inhibition of gap junction protein prevented microglial-glioma colony formation and microglia-mediated chemoresistance. CONCLUSIONS This study provides novel insights into how glioma cells acquire chemoresistance via microglia-mediated drug substance transferring, providing a new option for treating chemo-resistant brain tumors.
Collapse
Affiliation(s)
- Sheng-Yan Wu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Wen-Jui Yu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Ting-Yi Chien
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-An Ren
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chi-Shuo Chen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chi-Shiun Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan.
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, 30013, Taiwan.
- Frontier Research Center On Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
47
|
Liu H, Fu H, Yu C, Zhang N, Huang C, Lv L, Hu C, Chen F, Xiao Z, Zhang Z, Lu H, Yuan K. Transcriptional pausing induced by ionizing radiation enables the acquisition of radioresistance in nasopharyngeal carcinoma. J Mol Cell Biol 2024; 15:mjad044. [PMID: 37407287 PMCID: PMC10960568 DOI: 10.1093/jmcb/mjad044] [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: 11/25/2022] [Revised: 03/24/2023] [Accepted: 07/04/2023] [Indexed: 07/07/2023] Open
Abstract
Lesions on the DNA template can impact transcription via distinct regulatory pathways. Ionizing radiation (IR) as the mainstay modality for many malignancies elicits most of the cytotoxicity by inducing a variety of DNA damages in the genome. How the IR treatment alters the transcription cycle and whether it contributes to the development of radioresistance remain poorly understood. Here, we report an increase in the paused RNA polymerase II (RNAPII), as indicated by the phosphorylation at serine 5 residue of its C-terminal domain, in recurrent nasopharyngeal carcinoma (NPC) patient samples after IR treatment and cultured NPC cells developing IR resistance. Reducing the pool of paused RNAPII by either inhibiting TFIIH-associated CDK7 or stimulating the positive transcription elongation factor b, a CDK9-CycT1 heterodimer, attenuates IR resistance of NPC cells. Interestingly, the poly(ADP-ribosyl)ation of CycT1, which disrupts its phase separation, is elevated in the IR-resistant cells. Mutation of the major poly(ADP-ribosyl)ation sites of CycT1 decreases RNAPII pausing and restores IR sensitivity. Genome-wide chromatin immunoprecipitation followed by sequencing analyses reveal that several genes involved in radiation response and cell cycle control are subject to the regulation imposed by the paused RNAPII. Particularly, we identify the NIMA-related kinase NEK7 under such regulation as a new radioresistance factor, whose downregulation results in the increased chromosome instability, enabling the development of IR resistance. Overall, our results highlight a novel link between the alteration in the transcription cycle and the acquisition of IR resistance, opening up new opportunities to increase the efficacy of radiotherapy and thwart radioresistance in NPC.
Collapse
Affiliation(s)
- Honglu Liu
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Huanyi Fu
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Chunhong Yu
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Na Zhang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Canhua Huang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lu Lv
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410008, China
| | - Chunhong Hu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Fang Chen
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410008, China
| | - Zhiqiang Xiao
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhuohua Zhang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Huasong Lu
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Kai Yuan
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Biobank of Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
48
|
Batool A, Rashid W, Fatima K, Khan SU. Mechanisms of Cancer Resistance to Various Therapies. DRUG RESISTANCE IN CANCER: MECHANISMS AND STRATEGIES 2024:31-75. [DOI: 10.1007/978-981-97-1666-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
49
|
Negm A, Al-Faiyz YS, Riyadh SM, Sayed AR. Synthesis, DPPH Radical Scavenging, Cytotoxic Activity, and Apoptosis Induction Efficacy of Novel Thiazoles and Bis-thiazoles. Curr Org Synth 2024; 21:1081-1090. [PMID: 37936471 DOI: 10.2174/0115701794264504231017113027] [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: 06/15/2023] [Revised: 07/18/2023] [Accepted: 08/18/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Heterocyclic materials-containing thiazoles exhibited incredible importance in pharmaceutical chemistry and drug design due to their extensive biological properties. METHODS Synthesis of thiazoles and bis-thiazoles from the reaction of 2-((6-Nitrobenzo[ d][1,3]dioxol-5-yl)methylene)hydrazine-1-carbothioamide with hydrazonoyl chlorides in dioxane and in the existence of triethylamine as basic catalyst. The antioxidant, in vitro antiproliferative, and cytotoxicity efficacy of thiazoles and bis-thiazoles were measured. RESULTS In this work, novel series of 5-methyl-2-(2-(-(6-nitrobenzo[d][1,3]dioxol-5-yl)methylene) hydrazinyl)-4-(aryldiazenyl)thiazoles (4a-f) were prepared via the reaction of hydrazonoyl chlorides 2a-f with 2-((6-nitrobenzo[d][1,3]dioxol-5-yl)methylene)hydrazine-1-carbothioamide (1) in dioxane and employing triethylamine as basic catalyst. Following the same procedure, bisthiazoles (6, 8, and 10) have been synthesized by utilizing bis-hydrazonoyl chlorides (5, 7, and 9) and carbothioamide 1 in a molar ratio (1:2), respectively. The distinctive features in the structure of isolated products were elucidated by spectroscopic tools and elemental analyses. The antioxidant, in vitro anti-proliferative, cytotoxicity, and anti-cancer efficacy of thiazoles and bis-thiazoles were evaluated. Compounds 4d and 4f were the most potent antioxidant agents. Gene expression of apoptosis markers and fragmentation assay of DNA were assessed to explore the biochemical mechanism of synthesized products. Thiazoles significantly inhibited cell growth and proliferation more than bis-thiazoles. They induced apoptosis through induction of apoptotic gene expression P53 and downregulation of antiapoptotic gene expression Bcl-2. Moreover, they induced fragmentation of DNA in cancer cells, indicating that they could be employed as anticancer agents by inhibiting tumor growth and progression and can be considered effective compounds in the strategy of anti-cancer agents' discovery. CONCLUSION Synthesis, DPPH Radical Scavenging, Cytotoxic activity, and Apoptosis Induction Efficacy based on Novel Thiazoles and Bis-thiazoles.
Collapse
Affiliation(s)
- Amr Negm
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa, 31982, Saudi Arabia
- Chemistry Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | - Yasair S Al-Faiyz
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa, 31982, Saudi Arabia
| | - Sayed M Riyadh
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
- Department of Chemistry, Faculty of Science, Taibah University, Al-Madinah Almunawrah, 30002, Saudi Arabia
| | - Abdelwahed R Sayed
- Department of Chemistry, Faculty of Science, Beni-Suef University, Beni-suef, 62514, Egypt
| |
Collapse
|
50
|
Zihlif M, Hameduh T, Bulatova N, Hammad H. Alteration in the expression of the chemotherapy resistance‑related genes in response to chronic and acute hypoxia in pancreatic cancer. Biomed Rep 2023; 19:88. [PMID: 37901880 PMCID: PMC10603373 DOI: 10.3892/br.2023.1670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 09/08/2023] [Indexed: 10/31/2023] Open
Abstract
Pancreatic cancer is currently one of the least curable types of human cancer and remains a key health problem. One of the most important characteristics of pancreatic cancer is its ability to grow under hypoxic conditions. Hypoxia is associated with resistance of cancer cells to radiotherapy and chemotherapy. It is a major contributor to pancreatic cancer genetic instability, which local and systemic resistance that may result in poor clinical outcome. Accordingly, identifying gene expression changes in cancer resistance genes that occur under hypoxic conditions may identify a new therapeutic target. The aim of the present study was to explore the association between hypoxia and resistance to chemotherapy and determine the alteration in the expression of cancer resistance-related genes in the presence of hypoxia. Pancreatic cancer cells (PANC-1) were exposed to 8 h hypoxic episodes (<1% oxygen) three times/week for a total of 20 episodes (chronic hypoxia) or 72 h hypoxic episodes twice/week for a total of 10 episodes (acute hypoxia). The alterations in gene expression were examined using reverse transcription-quantitative PCR array compared with normoxic cells. Chemoresistance of hypoxic cells toward doxorubicin was assessed using MTT cell proliferation assay. Both chronic and acute hypoxia induced chemoresistance toward doxorubicin in PANC-1 pancreatic cancer cell line. The greatest changes occurred in estrogen Receptor Alpha Gene (ESR1) and ETS Like-1 protein (ELK1) pathways, in nucleic transcription factor Peroxisome proliferator-activated receptors (PPARs) and in a cell cycle inhibitor cyclin dependent kinase inhibitor 1A (CDKN1A). The present study demonstrated that exposing cells to prolonged hypoxia results in different gene expression changes involving pleotropic pathways that serve a role in inducing resistance in pancreatic cancer.
Collapse
Affiliation(s)
- Malek Zihlif
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Tareq Hameduh
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Nailya Bulatova
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Hana Hammad
- Department of Biology, School of Science, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|