1
|
Goyal A, Afzal M, Khan NH, Goyal K, Srinivasamurthy SK, Gupta G, Benod Kumar K, Ali H, Rana M, Wong LS, Kumarasamy V, Subramaniyan V. Targeting p53-p21 signaling to enhance mesenchymal stem cell regenerative potential. Regen Ther 2025; 29:352-363. [PMID: 40248767 PMCID: PMC12004386 DOI: 10.1016/j.reth.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 04/19/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are properties of self-renewal and differentiation potentials and thus are very appealing to regenerative medicine. Nevertheless, their therapeutic potential is frequently constrained by senescence, limited proliferation, and stress-induced apoptosis. The key role of the p53-p21 biology in MSC biology resides in safeguarding genomic stability while promoting senescence and limiting regenerative capacity upon over-activation demonstrated. This pathway is a key point for improving MSC function and exploiting the inherent limitations. Recent advances indicate that senescence can be delayed by targeting the p53-p21 signaling and improved MSC proliferation and differentiation capacity. PFT-α pharmacological agents transiently inhibit p53 from increasing proliferation and lineage-specific differentiation, while antioxidants such as hydrogen-rich saline and epigallocatechin 3 gallate (EGCG) suppress oxidative stress and attenuate p53 p21 signaling. Genetic tools like CRISPR-Cas9 and RNA interference also precisely modulate TP53 and CDKN1A expression to optimize MSC functionality. The interplay of p53-p21 with pathways like Wnt/β-catenin and MAPK further highlights opportunities for combinatorial therapies to enhance MSC resilience and regenerative outcomes. This review aims to offer a holistic view of how p53-p21 targeting can further the regenerative potential of MSCs, resolving senescence, proliferation, and stress resilience towards advanced therapeutics built on MSCs.
Collapse
Affiliation(s)
- Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Suresh Kumar Srinivasamurthy
- Department of Pharmacology, Ras Al Khaimah College of Medical Sciences, Ras Al Khaimah Medical & Health Sciences University, P.O. Box 11172, Ras Al Khaimah, United Arab Emirates
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - K. Benod Kumar
- Department of General Surgery, Consultant Head and Neck Surgical Oncology, Dr.D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai 71800, Malaysia
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Vetriselvan Subramaniyan
- Division of Pharmacology, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia
| |
Collapse
|
2
|
Zheng Z, Song Y. When HERG-caused LQT2 encounters antisense oligonucleotide: is exon 6 skipping therapy plausible? Front Pharmacol 2025; 16:1535259. [PMID: 40191427 PMCID: PMC11969117 DOI: 10.3389/fphar.2025.1535259] [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: 11/27/2024] [Accepted: 03/13/2025] [Indexed: 04/09/2025] Open
Abstract
The unique in-frame exon 6 of the HERG gene as a potential target for antisense oligonucleotide-mediated exon skipping therapy.
Collapse
Affiliation(s)
- Zequn Zheng
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yongfei Song
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
3
|
Ampolini EA, Jimenez-Sainz J, Long DT. The Development of ATM Inhibitors in Cancer Therapy. Target Oncol 2025; 20:281-297. [PMID: 40024979 PMCID: PMC11933189 DOI: 10.1007/s11523-025-01136-6] [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] [Accepted: 02/18/2025] [Indexed: 03/04/2025]
Abstract
The ataxia-telangiectasia mutated (ATM) protein kinase plays a critical role in activating the cellular response to DNA double-strand breaks and promoting homology-directed repair. ATM is frequently mutated in cancer, contributing to an accumulation of DNA damage that drives genomic instability. To exploit cancer cells' inherent vulnerability to DNA damage, various small molecule inhibitors have been developed that target ATM. ATM inhibitors have shown great versatility in preclinical studies and increasing use in the clinic. Here, we review the development of ATM inhibitors and their role in cancer therapy. We describe their limitations and the advances that have led to increases in both the number and diversity of active clinical trials targeting ATM. We also discuss ATM's role in personalized medicine and the current challenges to more widespread use of ATM inhibitors in the clinic.
Collapse
Affiliation(s)
- Elizabeth A Ampolini
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Judit Jimenez-Sainz
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - David T Long
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
4
|
Li H, Yang Y, Li B, Yang J, Liu P, Gao Y, Zhang M, Ning G. Comprehensive Analysis Reveals the Potential Diagnostic Value of Biomarkers Associated With Aging and Circadian Rhythm in Knee Osteoarthritis. Orthop Surg 2025; 17:922-938. [PMID: 39846237 PMCID: PMC11872380 DOI: 10.1111/os.14370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/24/2025] Open
Abstract
OBJECTIVE Knee osteoarthritis (KOA) is characterized by structural changes. Aging is a major risk factor for KOA. Therefore, the objective of this study was to examine the role of genes related to aging and circadian rhythms in KOA. METHODS This study identified differentially expressed genes (DEGs) by comparing gene expression levels between normal and KOA samples from the GEO database. Subsequently, we intersected the DEGs with aging-related circadian rhythm genes to obtain a set of aging-associated circadian rhythm genes differentially expressed in KOA. Next, we conducted Mendelian randomization (MR) analysis, using the differentially expressed aging-related circadian rhythm genes in KOA as the exposure factors, their SNPs as instrumental variables, and KOA as the outcome event, to explore the causal relationship between these genes and KOA. We then performed Gene Set Enrichment Analysis (GSEA) to investigate the pathways associated with the selected biomarkers, conducted immune infiltration analysis, built a competing endogenous RNA (ceRNA) network, and performed molecular docking studies. Additionally, the findings and functional roles of the biomarkers were further validated through experiments on human cartilage tissue and cell models. RESULTS A total of 75 differentially expressed aging-circadian rhythm related genes between the normal group and the KOA group were identified by difference analysis, primarily enriched in the circadian rhythm pathway. Two biomarkers (PFKFB4 and DDIT4) were screened by MR analysis. Then, immune infiltration analysis showed significant differences in three types of immune cells (resting dendritic cells, resting mast cells, and M2 macrophages), between the normal and KOA groups. Drug prediction and molecular docking results indicated stable binding of PFKFB4 to estradiol and bisphenol_A, while DDIT4 binds stably to nortriptyline and trimipramine. Finally, cell lines with stable expression of the biomarkers were established by lentiviral infection and resistance screening, Gene expression was significantly elevated in overexpressing cells of PFKFB4 and DDIT4 and reversed the proliferation and migration ability of cells after IL-1β treatment. CONCLUSIONS Two diagnostic and therapeutic biomarkers associated with aging-circadian rhythm in KOA were identified. Functional analysis, molecular mechanism exploration, and experimental validation further elucidated their roles in KOA, offering novel perspectives for the prevention and treatment of the disease.
Collapse
Affiliation(s)
- Hao Li
- Department of OrthopedicsTianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal CordTianjinChina
| | - Yuze Yang
- Department of OrthopedicsThe Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairTaiyuanChina
| | - Bo Li
- Department of OrthopedicsTianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal CordTianjinChina
| | - Jiaju Yang
- Department of OrthopedicsThe Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairTaiyuanChina
| | - Pengyu Liu
- Department of OrthopedicsThe Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairTaiyuanChina
| | - Yuanpeng Gao
- Department of OrthopedicsThe Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairTaiyuanChina
| | - Min Zhang
- Department of OrthopedicsThe Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairTaiyuanChina
| | - Guangzhi Ning
- Department of OrthopedicsTianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal CordTianjinChina
| |
Collapse
|
5
|
Islam SM, Hasan M, Alam J, Dey A, Molineaux D. In Silico Screening, Molecular Dynamics Simulation and Binding Free Energy Identify Single-Point Mutations That Destabilize p53 and Reduce Binding to DNA. Proteins 2025; 93:498-514. [PMID: 39264222 PMCID: PMC11695177 DOI: 10.1002/prot.26747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/09/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Considering p53's pivotal role as a tumor suppressor protein, proactive identification and characterization of potentially harmful p53 mutations are crucial before they appear in the population. To address this, four computational prediction tools-SIFT, Polyphen-2, PhD-SNP, and MutPred2-utilizing sequence-based and machine-learning algorithms, were employed to identify potentially deleterious p53 nsSNPs (nonsynonymous single nucleotide polymorphisms) that may impact p53 structure, dynamics, and binding with DNA. These computational methods identified three variants, namely, C141Y, C238S, and L265P, as detrimental to p53 stability. Furthermore, molecular dynamics (MD) simulations revealed that all three variants exhibited heightened structural flexibility compared to the native protein, especially the C141Y and L265P mutations. Consequently, due to the altered structure of mutant p53, the DNA-binding affinity of all three variants decreased by approximately 1.8 to 9.7 times compared to wild-type p53 binding with DNA (14 μM). Notably, the L265P mutation exhibited an approximately ten-fold greater reduction in binding affinity. Consequently, the presence of the L265P mutation in p53 could pose a substantial risk to humans. Given that p53 regulates abnormal tumor growth, this research carries significant implications for surveillance efforts and the development of anticancer therapies.
Collapse
Affiliation(s)
- Shahidul M. Islam
- Department of Chemistry, Delaware State University, Dover, DE 19901, USA
| | - Mehedi Hasan
- Department of Chemistry, Delaware State University, Dover, DE 19901, USA
| | - Jahidul Alam
- Department of Molecular Biology and Biotechnology, Queen’s University Belfast, Northern Ireland, BT7 1NN, United Kingdom
| | - Anonya Dey
- Department of Genetic Engineering and Biotechnology, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Dylan Molineaux
- Department of Chemistry, Delaware State University, Dover, DE 19901, USA
| |
Collapse
|
6
|
Brane A, Sutko M, Tollefsbol TO. p21 Promoter Methylation Is Vital for the Anticancer Activity of Withaferin A. Int J Mol Sci 2025; 26:1210. [PMID: 39940977 PMCID: PMC11818515 DOI: 10.3390/ijms26031210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/23/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
Breast cancer (BC) is a widespread malignancy that affects the lives of millions of women each year, and its resulting financial and healthcare hardships cannot be overstated. These issues, in combination with side effects and obstacles associated with the current standard of care, generate considerable interest in new potential targets for treatment as well as means for BC prevention. One potential preventive compound is Withaferin A (WFA), a traditional medicinal compound found in winter cherries. WFA has shown promise as an anticancer agent and is thought to act primarily through its effects on the epigenome, including, in particular, the methylome. However, the relative importance of specific genes' methylation states to WFA function remains unclear. To address this, we utilized human BC cell lines in combination with CRISPR-dCas9 fused to DNA methylation modifiers (i.e., epigenetic editors) to elucidate the importance of specific genes' promoter methylation states to WFA function and cancer cell viability. We found that targeted demethylation of promoters of the tumor suppressors p21 and p53 within MDA-MB-231/MCF7 cells resulted in around 1.7×/1.5× and 1.2×/1.3× increases in expression, respectively. Targeted methylation of the promoter of the oncogene CCND1 within MDA-MB-231/MCF7 cells resulted in 0.5×/0.8× decreases in gene expression. These changes to p21, p53, and CCND1 were also associated with decreases in cell viability of around 25%/50%, 5%/35%, and 12%/16%, respectively, for MDA-MB-231/MCF7 cells. When given in combination with WFA in both p53 mutant and wild type cells, we discovered that targeted methylation of the p21 promoter was able to modulate the anticancer effects of WFA, while targeted methylation or demethylation of the promoters of p53 and CCND1 had no significant effect on viability decreases from WFA treatment. Taken together, these results indicate that p21, p53, and CCND1 may be important targets for future in vivo studies that may lead to epigenetic editing therapies and that WFA may have utility in the prevention of BC through its effect on p21 promoter methylation independent of p53 function.
Collapse
Affiliation(s)
- Andrew Brane
- Department of Biology, University of Alabama at Birmingham, 3100 Science & Engineering Complex—East Science Hall, 902 14th Street South, Birmingham, AL 35205, USA
| | - Madeline Sutko
- Department of Biology, University of Alabama at Birmingham, 3100 Science & Engineering Complex—East Science Hall, 902 14th Street South, Birmingham, AL 35205, USA
| | - Trygve O. Tollefsbol
- Department of Biology, University of Alabama at Birmingham, 3100 Science & Engineering Complex—East Science Hall, 902 14th Street South, Birmingham, AL 35205, USA
- Comprehensive Cancer Center, University of Alabama at Birmingham, 1802 6th Avenue South, Birmingham, AL 35294, USA
- Integrative Center for Aging Research, University of Alabama at Birmingham, 933 19th Street South, Birmingham, AL 35294, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, 1675 University Blvd, Birmingham, AL 35294, USA
- Comprehensive Diabetes Center, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL 35294, USA
| |
Collapse
|
7
|
Pan Y, Chen H, Fu J, Zhang J, Wang P, Chen R, Geng S, Che J, Dong X, Zhou Y, Huang W. Discovery of N-Benzylpiperidinol derivatives as USP7 inhibitors against Hematology. Bioorg Chem 2024; 153:107807. [PMID: 39293304 DOI: 10.1016/j.bioorg.2024.107807] [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: 07/14/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
USP7 has been recognized as a potential target for the treatment of hematologic malignancies by stabilizing multiple cancer-relevant proteins. Nevertheless, drug-like USP7 inhibitors are still lacking. Herein, compound J21 (USP7 IC50: 41.35 ± 2.16 nM) was discovered based on the structure of L55 and its co-crystal complex with USP7. Additionally, J21 exhibited greater metabolic stability (T1/2: 1.25 h, Cmax: 394.1 ± 48.3 ng/mL, and AUC0-t: 597.8 ± 44.8 ng/mL∙h) compared to L55. These findings may further pave the way for the development of USP7 inhibitors for the treatment of hematologic malignancies.
Collapse
Affiliation(s)
- Youlu Pan
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou 310058, China
| | - Haifeng Chen
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jingfeng Fu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medical, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Guangdong 528400, China
| | - Jingyu Zhang
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Peipei Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medical, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Guangdong 528400, China
| | - Runmei Chen
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou 310058, China
| | - Shuangshuang Geng
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jinxin Che
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yubo Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medical, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Guangdong 528400, China.
| | - Wenhai Huang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou 310058, China.
| |
Collapse
|
8
|
Wei Q, Yang Y, Li C, Wang H. ZC3H13-induced the m6A modification of hsa_circ_0081723 promotes cervical cancer progression via AMPK/p53 pathway. J Obstet Gynaecol Res 2024; 50:2286-2298. [PMID: 39476847 DOI: 10.1111/jog.16140] [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: 07/09/2024] [Accepted: 10/22/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND N6-methyladenosine (m6A) modification and circular RNAs (circRNAs) have been confirmed to participate in cervical cancer (CC) progression. However, the function of a novel circRNA, hsa_circ_0081723, has not yet been explored in CC. Therefore, this study aimed to investigate the potential role of hsa_circ_0081723 and its m6A modification in CC. METHODS The hsa_circ_0081723 and ZC3H13 expressions were examined by qRT-PCR in the CC tissues, and their prognostic significance was evaluated via Kaplan-Meier Plotter. The role of hsa_circ_0081723 in CC progression was checked by loss-of-function assays. The relative protein levels of AMPK/p53 pathway were determined by western blotting. The interactions of hsa_circ_0081723 and ZC3H13 were verified via MeRIP and RNA stability assays. RESULTS The hsa_circ_0081723 expression was elevated in CC samples, and its higher levels indicated high histological grade, high FIGO stage, poor differentiation, and poor prognosis. Functionally, silencing hsa_circ_0081723 impaired the malignant behavior of CC cells and enhanced the protein levels of key molecules of the AMPK signaling pathway. Moreover, ZC3H13 was also elevated in CC samples and demonstrated a positive association with hsa_circ_0081723. The relative enrichment of hsa_circ_0081723 m6A and its stability were enhanced in ZC3H13 overexpressed CC cells. Mechanically, ZC3H13 overexpression partially reversed the antitumor effects caused by hsa_circ_0081723 knockdown in CC cells. CONCLUSIONS This study innovatively demonstrates that ZC3H13-mediated m6A modification of hsa_circ_0081723 promotes CC progression by modulating AMPK/p53 pathway. Our findings may contribute to the understanding of the molecular mechanisms underlying CC and offer potential therapeutic targets for clinical treatment.
Collapse
Affiliation(s)
- Qiong Wei
- Department of Obstetrics and Gynecology, Wuhan Third Hospital, Wuhan, Hubei, China
| | - Yi Yang
- Department of Obstetrics and Gynecology, Wuhan Third Hospital, Wuhan, Hubei, China
| | - Chun Li
- Department of Obstetrics and Gynecology, Wuhan Third Hospital, Wuhan, Hubei, China
| | - Huimin Wang
- Department of Obstetrics and Gynecology, Wuhan Third Hospital, Wuhan, Hubei, China
| |
Collapse
|
9
|
Lan T, He S, Luo X, Pi Z, Lai W, Jiang C, Gan J, Wei S, Wu Z, Yun C, Leng J, Li C. Disruption of NADPH homeostasis by total flavonoids from Adinandra nitida Merr. ex Li leaves triggers ROS-dependent p53 activation leading to apoptosis in non-small cell lung cancer cells. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118340. [PMID: 38762212 DOI: 10.1016/j.jep.2024.118340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/03/2024] [Accepted: 05/11/2024] [Indexed: 05/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Adinandra nitida Merr. ex Li leaves serve as a herbal tea and hold a significant role in traditional Chinese medicine, being applied to assist in tumor treatment. Flavonoids present the primary bioactive constituents in Adinandra nitida Merr. ex Li leaves. AIM OF THE STUDY To explore the potential of total flavonoids from Adinandra nitida Merr. ex Li Leaves (TFAN) in inhibiting non-small cell lung cancer (NSCLC) and further elucidate the underlying mechanisms. MATERIALS AND METHODS Human NSCLC cell lines and normal lung cell line were employed to assess the impact of TFAN (0-160 μg/mL for 24, 28 and 72 h) on cell proliferation in vitro. Immunofluorescence (IF) staining gauged p53 expression changes in NSCLC cells under TFAN present condition (150 μg/mL for 24 h). In vivo study utilized NSCLC cell derived xenograft tumors in nude mice, administering TFAN orally (200 and 400 mg/kg) for 14 days. Immunohistochemistry assessed Cleaved Caspase 3 expression change in A549 xenograft tumors treated with TFAN (400 mg/kg for 14 days). RNA-seq and KEGG analysis identified gene expression changes and enriched processes in A549 xenograft tumors treated with TFAN. CM-H2DCFDA and metabolomics assessed ROS level and GSH/GSSG pool changes in A549 cells under TFAN present condition. Cell viability assay and IF staining assessed A549 cell proliferation and p53 expression changes under H2O2-induced oxidative stress (0-40 μM for 24 h) and TFAN present conditions. GSEA and N-Acetyl-L-cysteine (NAC) rescue (0-1 μM for 24 h) analyzed the impact of TFAN on GSH de novo synthesis. NADPH/NADP+ pool measurement and NADPH rescue (0-10 μM for 24 h) analyzed the impact of TFAN on GSH salvage synthesis. GC-FID and HPLC-MS were utilized to detect ethanol and ethyl acetate residues, and to characterize the chemical constituents in TFAN, respectively. The total flavonoid content of TFAN was determined using a 330 nm wavelength. RESULTS TFAN significantly inhibited A549 cells (wild-type p53) but not NCI-H1299 cells (p53-deficient), NCI-H596 cells (p53-mutant) or BEAS-2B in vitro. IF staining validated p53 genotype for the cell lines and revealed an increase in p53 expression in A549 cells after TFAN treatment. In vivo, TFAN selectively inhibited A549 xenograft tumor growth without discernible toxicity, inducing apoptosis evidenced by Cleaved Caspase 3 upregulation. RNA-seq and KEGG analysis suggested ROS biosynthesis was involved in TFAN-induced p53 activation in A549 cells. Elevated ROS level in TFAN-treated A549 cells were observed. Moreover, TFAN sensitized A549 cells to H2O2-induced oxidative stress, with higher p53 expression. Additionally, A549 cells compensated with GSH de novo synthesis under TFAN present condition, confirmed by GSEA and NAC rescue experiment. TFAN disrupted NADPH homeostasis to impair GSH salvage biosynthesis, supported by NADPH/NADP+ change and NADPH rescue experiment. The chemical constituents of TFAN, with acceptable limits for ethanol and ethyl acetate residues and a total flavonoid content of 68.87%, included Catechin, Epicatechin, Quercitroside, Camellianin A, and Apigenin. CONCLUSION The disruption of NADPH homeostasis by TFAN triggers ROS-dependent p53 activation that leads to apoptotic cell death, ultimately suppressing NSCLC growth. These findings offer potential therapeutic implications of Adinandra nitida Merr. ex Li leaves in combating NSCLC.
Collapse
Affiliation(s)
- Taijin Lan
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; Key Laboratory of Integrative Translational Medicine of Guangxi High Incidence Infectious Diseases, Nanning 530200, China; School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Songhua He
- Guangxi Institute for Food and Drug Control, Nanning 530021, China
| | - Xuefei Luo
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Zhenyu Pi
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Weihui Lai
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Chunhui Jiang
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Jun Gan
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Suyun Wei
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Zhanshuai Wu
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Chenxia Yun
- Key Laboratory of Integrative Translational Medicine of Guangxi High Incidence Infectious Diseases, Nanning 530200, China.
| | - Jing Leng
- Key Laboratory of Integrative Translational Medicine of Guangxi High Incidence Infectious Diseases, Nanning 530200, China.
| | - Changlong Li
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
10
|
Temaj G, Chichiarelli S, Telkoparan-Akillilar P, Saha S, Nuhii N, Hadziselimovic R, Saso L. P53: A key player in diverse cellular processes including nuclear stress and ribosome biogenesis, highlighting potential therapeutic compounds. Biochem Pharmacol 2024; 226:116332. [PMID: 38830426 DOI: 10.1016/j.bcp.2024.116332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/05/2024]
Abstract
The tumor suppressor proteins are key transcription factors involved in the regulation of various cellular processes, such as apoptosis, DNA repair, cell cycle, senescence, and metabolism. The tumor suppressor protein p53 responds to different type of stress signaling, such as hypoxia, DNA damage, nutrient deprivation, oncogene activation, by activating or repressing the expression of different genes that target processes mentioned earlier. p53 has the ability to modulate the activity of many other proteins and signaling pathway through protein-protein interaction, post-translational modifications, or non-coding RNAs. In many cancers the p53 is found to be mutated or inactivated, resulting in the loss of its tumor suppressor function and acquisition of new oncogenic properties. The tumor suppressor protein p53 also plays a role in the development of other metabolic disorders such as diabetes, obesity, and fatty liver disease. In this review, we will summarize the current data and knowledge on the molecular mechanisms and the functions of p53 in different pathways and processes at the cellular level and discuss the its implications for human health and disease.
Collapse
Affiliation(s)
- Gazmend Temaj
- Faculty of Pharmacy, College UBT, 10000 Prishtina, Kosovo.
| | - Silvia Chichiarelli
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, 00185 Rome, Italy.
| | | | - Sarmistha Saha
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura 00185, Uttar Pradesh, India.
| | - Nexhibe Nuhii
- Department of Pharmacy, Faculty of Medical Sciences, State University of Tetovo, 1200 Tetovo, Macedonia.
| | - Rifat Hadziselimovic
- Faculty of Science, University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina.
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", La Sapienza University, 00185 Rome, Italy.
| |
Collapse
|
11
|
Musa S, Amara N, Selawi A, Wang J, Marchini C, Agbarya A, Mahajna J. Overcoming Chemoresistance in Cancer: The Promise of Crizotinib. Cancers (Basel) 2024; 16:2479. [PMID: 39001541 PMCID: PMC11240740 DOI: 10.3390/cancers16132479] [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: 06/11/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Chemoresistance is a major obstacle in cancer treatment, often leading to disease progression and poor outcomes. It arises through various mechanisms such as genetic mutations, drug efflux pumps, enhanced DNA repair, and changes in the tumor microenvironment. These processes allow cancer cells to survive despite chemotherapy, underscoring the need for new strategies to overcome resistance and improve treatment efficacy. Crizotinib, a first-generation multi-target kinase inhibitor, is approved by the FDA for the treatment of ALK-positive or ROS1-positive non-small cell lung cancer (NSCLC), refractory inflammatory (ALK)-positive myofibroblastic tumors (IMTs) and relapsed/refractory ALK-positive anaplastic large cell lymphoma (ALCL). Crizotinib exists in two enantiomeric forms: (R)-crizotinib and its mirror image, (S)-crizotinib. It is assumed that the R-isomer is responsible for the carrying out various processes reviewed here The S-isomer, on the other hand, shows a strong inhibition of MTH1, an enzyme important for DNA repair mechanisms. Studies have shown that crizotinib is an effective multi-kinase inhibitor targeting various kinases such as c-Met, native/T315I Bcr/Abl, and JAK2. Its mechanism of action involves the competitive inhibition of ATP binding and allosteric inhibition, particularly at Bcr/Abl. Crizotinib showed synergistic effects when combined with the poly ADP ribose polymerase inhibitor (PARP), especially in ovarian cancer harboring BRCA gene mutations. In addition, crizotinib targets a critical vulnerability in many p53-mutated cancers. Unlike its wild-type counterpart, the p53 mutant promotes cancer cell survival. Crizotinib can cause the degradation of the p53 mutant, sensitizing these cancer cells to DNA-damaging substances and triggering apoptosis. Interestingly, other reports demonstrated that crizotinib exhibits anti-bacterial activity, targeting Gram-positive bacteria. Also, it is active against drug-resistant strains. In summary, crizotinib exerts anti-tumor effects through several mechanisms, including the inhibition of kinases and the restoration of drug sensitivity. The potential of crizotinib in combination therapies is emphasized, particularly in cancers with a high prevalence of the p53 mutant, such as triple-negative breast cancer (TNBC) and high-grade serous ovarian cancer (HGSOC).
Collapse
Affiliation(s)
- Sanaa Musa
- Department of Nutrition and Natural Products, Migal-Galilee Research Institute, Kiryat Shmona 11016, Israel
- Department of Biotechnology, Tel-Hai College, Kiryat Shmona 11016, Israel
| | - Noor Amara
- Department of Nutrition and Natural Products, Migal-Galilee Research Institute, Kiryat Shmona 11016, Israel
- Department of Biotechnology, Tel-Hai College, Kiryat Shmona 11016, Israel
| | - Adan Selawi
- Department of Nutrition and Natural Products, Migal-Galilee Research Institute, Kiryat Shmona 11016, Israel
- Department of Biotechnology, Tel-Hai College, Kiryat Shmona 11016, Israel
| | - Junbiao Wang
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Cristina Marchini
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Abed Agbarya
- Oncology Department, Bnai Zion MC, Haifa 31048, Israel
| | - Jamal Mahajna
- Department of Nutrition and Natural Products, Migal-Galilee Research Institute, Kiryat Shmona 11016, Israel
- Department of Biotechnology, Tel-Hai College, Kiryat Shmona 11016, Israel
| |
Collapse
|
12
|
Holý P, Hlaváč V, Šeborová K, Šůsová S, Tesařová T, Rob L, Hruda M, Bouda J, Bartáková A, Mrhalová M, Kopečková K, Al Obeed Allah M, Špaček J, Sedláková I, Souček P, Václavíková R. Targeted DNA sequencing of high-grade serous ovarian carcinoma reveals association of TP53 mutations with platinum resistance when combined with gene expression. Int J Cancer 2024; 155:104-116. [PMID: 38447012 DOI: 10.1002/ijc.34908] [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: 11/07/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 03/08/2024]
Abstract
High-grade serous ovarian carcinoma (HGSC) is the most common subtype of ovarian cancer and is among the most fatal gynecological malignancies worldwide, due to late diagnosis at advanced stages and frequent therapy resistance. In 47 HGSC patients, we assessed somatic and germline genetic variability of a custom panel of 144 known or suspected HGSC-related genes by high-coverage targeted DNA sequencing to identify the genetic determinants associated with resistance to platinum-based therapy. In the germline, the most mutated genes were DNAH14 (17%), RAD51B (17%), CFTR (13%), BRCA1 (11%), and RAD51 (11%). Somatically, the most mutated gene was TP53 (98%), followed by CSMD1/2/3 (19/19/36%), and CFTR (23%). Results were compared with those from whole exome sequencing of a similar set of 35 HGSC patients. Somatic variants in TP53 were also validated using GENIE data of 1287 HGSC samples. Our approach showed increased prevalence of high impact somatic and germline mutations, especially those affecting splice sites of TP53, compared to validation datasets. Furthermore, nonsense TP53 somatic mutations were negatively associated with patient survival. Elevated TP53 transcript levels were associated with platinum resistance and presence of TP53 missense mutations, while decreased TP53 levels were found in tumors carrying mutations with predicted high impact, which was confirmed in The Cancer Genome Atlas data (n = 260). Targeted DNA sequencing of TP53 combined with transcript quantification may contribute to the concept of precision oncology of HGSC. Future studies should explore targeting the p53 pathway based on specific mutation types and co-analyze the expression and mutational profiles of other key cancer genes.
Collapse
Affiliation(s)
- Petr Holý
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Viktor Hlaváč
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Karolína Šeborová
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Simona Šůsová
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Tereza Tesařová
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Lukáš Rob
- Department of Gynecology and Obstetrics, Third Faculty of Medicine and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Martin Hruda
- Department of Gynecology and Obstetrics, Third Faculty of Medicine and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Jiří Bouda
- Department of Gynecology and Obstetrics, University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Alena Bartáková
- Department of Gynecology and Obstetrics, University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Marcela Mrhalová
- Department of Pathology and Molecular Medicine, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kateřina Kopečková
- Department of Oncology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Mohammad Al Obeed Allah
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Jiří Špaček
- University Hospital Hradec Králové, Hradec Kralove, Czech Republic
| | - Iva Sedláková
- University Hospital Hradec Králové, Hradec Kralove, Czech Republic
| | - Pavel Souček
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Radka Václavíková
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| |
Collapse
|
13
|
Zhang S, El-Deiry WS. Transfected SARS-CoV-2 spike DNA for mammalian cell expression inhibits p53 activation of p21(WAF1), TRAIL Death Receptor DR5 and MDM2 proteins in cancer cells and increases cancer cell viability after chemotherapy exposure. Oncotarget 2024; 15:275-284. [PMID: 38709242 PMCID: PMC11073320 DOI: 10.18632/oncotarget.28582] [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: 04/16/2024] [Accepted: 04/30/2024] [Indexed: 05/07/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and COVID-19 infection has led to worsened outcomes for patients with cancer. SARS-CoV-2 spike protein mediates host cell infection and cell-cell fusion that causes stabilization of tumor suppressor p53 protein. In-silico analysis previously suggested that SARS-CoV-2 spike interacts with p53 directly but this putative interaction has not been demonstrated in cells. We examined the interaction between SARS-CoV-2 spike, p53 and MDM2 (E3 ligase, which mediates p53 degradation) in cancer cells using an immunoprecipitation assay. We observed that SARS-CoV-2 spike protein interrupts p53-MDM2 protein interaction but did not detect SARS-CoV-2 spike bound with p53 protein in the cancer cells. We further observed that SARS-CoV-2 spike suppresses p53 transcriptional activity in cancer cells including after nutlin exposure of wild-type p53-, spike-expressing tumor cells and inhibits chemotherapy-induced p53 gene activation of p21(WAF1), TRAIL Death Receptor DR5 and MDM2. The suppressive effect of SARS-CoV-2 spike on p53-dependent gene activation provides a potential molecular mechanism by which SARS-CoV-2 infection may impact tumorigenesis, tumor progression and chemotherapy sensitivity. In fact, cisplatin-treated tumor cells expressing spike were found to have increased cell viability as compared to control cells. Further observations on γ-H2AX expression in spike-expressing cells treated with cisplatin may indicate altered DNA damage sensing in the DNA damage response pathway. The preliminary observations reported here warrant further studies to unravel the impact of SARS-CoV-2 and its various encoded proteins including spike on pathways of tumorigenesis and response to cancer therapeutics. More efforts should be directed at studying the effects of the SARS-CoV-2 spike and other viral proteins on host DNA damage sensing, response and repair mechanisms. A goal would be to understand the structural basis for maximal anti-viral immunity while minimizing suppression of host defenses including the p53 DNA damage response and tumor suppression pathway. Such directions are relevant and important including not only in the context of viral infection and mRNA vaccines in general but also for patients with cancer who may be receiving cytotoxic or other cancer treatments.
Collapse
Affiliation(s)
- Shengliang Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI 02912, USA
- Legorreta Cancer Center at Brown University, Providence, RI 02912, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI 02912, USA
- Legorreta Cancer Center at Brown University, Providence, RI 02912, USA
- Hematology/Oncology Division, Department of Medicine, Lifespan Health System and Brown University, Providence, RI 02912, USA
| |
Collapse
|
14
|
Kamath D, Iwakuma T, Bossmann SH. Therapeutic potential of combating cancer by restoring wild-type p53 through mRNA nanodelivery. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 56:102732. [PMID: 38199451 PMCID: PMC11108594 DOI: 10.1016/j.nano.2024.102732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/27/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024]
Abstract
Among the tumor suppressor genes, TP53 is the most frequently mutated in human cancers, and most mutations are missense mutations causing production of mutant p53 (mutp53) proteins. TP53 mutations not only results in loss of function (LOH) as a transcription factor and a tumor suppressor, but also gain wild-type p53 (WTp53)-independent oncogenic functions that enhance cancer metastasis and progression (Yamamoto and Iwakuma, 2018; Zhang et al., 2022). TP53 has extensively been studied as a therapeutic target as well as for drug development and therapies, however with limited success. Achieving targeted therapies for restoration of WTp53 function and depletion or repair of mutant p53 (mutp53) will have far reaching implication in cancer treatment and therapies. This review briefly discusses the role of p53 mutation in cancer and the therapeutic potential of restoring WTp53 through the advances in mRNA nanomedicine.
Collapse
Affiliation(s)
- Divya Kamath
- The University of Kansas Medical Center, Department of Cancer Biology, 3901 Rainbow Blvd, mailstop 1071, 66160 Kansas City, KS, USA.
| | - Tomoo Iwakuma
- Children's Mercy Hospital, Adele Hall Campus, 2401 Gillham Rd, Kansas City, MO 64108, USA.
| | - Stefan H Bossmann
- The University of Kansas Medical Center, Department of Cancer Biology, 3901 Rainbow Blvd, mailstop 1071, 66160 Kansas City, KS, USA.
| |
Collapse
|
15
|
Chatterjee T, Guha D, Dhar J, Saha T, Paul D, Sa G, Chakrabarti P. Adenosine dialdehyde, a methyltransferase inhibitor, induces colorectal cancer cells apoptosis by regulating PIMT:p53 interaction. Biochem Biophys Res Commun 2023; 684:149134. [PMID: 37871521 DOI: 10.1016/j.bbrc.2023.149134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 10/03/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023]
Abstract
Post-translational modification (PTM) is important in controlling many biological processes by changing the structure and function of a protein. Protein methylation is an important PTM, and the role of methyltransferases has been implicated in numerous cellular functions. Protein L-isoaspartyl methyltransferase (PIMT) is ubiquitously expressed in almost all organisms and govern important cellular processes including apoptosis. Among other functions, PIMT has also been identified as a potent oncogene because it destabilizes the structure of the tumor suppressor p53 via methylation at the transactivation domain. In the present study we identified that out of the three methyltransferase inhibitors tested, namely, S-adenosyl-l-homocysteine (AdoHcy), adenosine and adenosine dialdehyde (AdOx), only AdOx augments p53 expression by destabilizing PIMT structure, as evident from far-UV CD. The effect of the inhibitors, AdOx in particular, to the structure of PIMT, and the binding of PIMT to the p53 transactivation domain have been investigated by docking and molecular dynamics simulations. AdOx significantly increases p53 accumulation and nuclear translocation in colon cancer cells, triggering the p53-mediated apoptotic pathway. To better understand the molecular mechanisms underlying p53 accumulation in colon cancer cells, we observed that the level of PIMT is considerably lower in AdOx-treated cells, reducing its association with p53, which stabilized p53. p53 then transactivated BAX, increasing the BAX: BCL-2 ratio and causing colon cancer cell death.
Collapse
Affiliation(s)
- Tanaya Chatterjee
- Department of Biochemistry, Bose Institute, P1/12, CIT Scheme VII M, Kolkata, 700054, India.
| | - Deblina Guha
- Division of Molecular Medicine, Bose Institute, P1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Jesmita Dhar
- Division of Molecular Medicine, Bose Institute, P1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Taniya Saha
- Division of Molecular Medicine, Bose Institute, P1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Debamita Paul
- Department of Biochemistry, Bose Institute, P1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Gaurisankar Sa
- Division of Molecular Medicine, Bose Institute, P1/12, CIT Scheme VII M, Kolkata, 700054, India.
| | - Pinak Chakrabarti
- Department of Biochemistry, Bose Institute, P1/12, CIT Scheme VII M, Kolkata, 700054, India
| |
Collapse
|
16
|
Heczko L, Hlaváč V, Holý P, Dvořák P, Liška V, Vyčítal O, Fiala O, Souček P. Prognostic potential of whole exome sequencing in the clinical management of metachronous colorectal cancer liver metastases. Cancer Cell Int 2023; 23:295. [PMID: 38008721 PMCID: PMC10676609 DOI: 10.1186/s12935-023-03135-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/09/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND Colorectal cancer is a highly prevalent and deadly. The most common metastatic site is the liver. We performed a whole exome sequencing analysis of a series of metachronous colorectal cancer liver metastases (mCLM) and matched non-malignant liver tissues to investigate the genomic profile of mCLM and explore associations with the patients' prognosis and therapeutic modalities. METHODS DNA samples from mCLM and non-malignant liver tissue pairs (n = 41) were sequenced using whole exome target enrichment and their germline and somatic genetic variability, copy number variations, and mutational signatures were assessed for associations with relapse-free (RFS) and overall survival (OS). RESULTS Our genetic analysis could stratify all patients into existing targeted therapeutic regimens. The most commonly mutated genes in mCLM were TP53, APC, and KRAS together with PIK3CA and several passenger genes like ABCA13, FAT4, PCLO, and UNC80. Patients with somatic alterations in genes from homologous recombination repair, Notch, and Hedgehog pathways had significantly prolonged RFS, while those with altered MYC pathway genes had poor RFS. Additionally, alterations in the JAK-STAT pathway were prognostic of longer OS. Patients bearing somatic variants in VIPR2 had significantly shorter OS and those with alterations in MUC16 prolonged OS. Carriage of the KRAS-12D variant was associated with shortened survival in our and external datasets. On the other hand, tumor mutation burden, mismatch repair deficiency, microsatellite instability, mutational signatures, or copy number variation in mCLM had no prognostic value. CONCLUSIONS The results encourage further molecular profiling for personalized treatment of colorectal cancer liver metastases discerning metachronous from synchronous scenarios.
Collapse
Affiliation(s)
- Lucie Heczko
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, 306 05, Czech Republic
| | - Viktor Hlaváč
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, 306 05, Czech Republic
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
| | - Petr Holý
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, 306 05, Czech Republic
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
| | - Pavel Dvořák
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, 306 05, Czech Republic
- Department of Biology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Václav Liška
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, 306 05, Czech Republic
- Department of Surgery, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Ondřej Vyčítal
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, 306 05, Czech Republic
- Department of Surgery, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Ondřej Fiala
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, 306 05, Czech Republic
- Department of Oncology and Radiotherapeutics, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Souček
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, 306 05, Czech Republic.
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic.
| |
Collapse
|
17
|
Carlsen L, Zhang S, Tian X, De La Cruz A, George A, Arnoff TE, El-Deiry WS. The role of p53 in anti-tumor immunity and response to immunotherapy. Front Mol Biosci 2023; 10:1148389. [PMID: 37602328 PMCID: PMC10434531 DOI: 10.3389/fmolb.2023.1148389] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 07/04/2023] [Indexed: 08/22/2023] Open
Abstract
p53 is a transcription factor that regulates the expression of genes involved in tumor suppression. p53 mutations mediate tumorigenesis and occur in approximately 50% of human cancers. p53 regulates hundreds of target genes that induce various cell fates including apoptosis, cell cycle arrest, and DNA damage repair. p53 also plays an important role in anti-tumor immunity by regulating TRAIL, DR5, TLRs, Fas, PKR, ULBP1/2, and CCL2; T-cell inhibitory ligand PD-L1; pro-inflammatory cytokines; immune cell activation state; and antigen presentation. Genetic alteration of p53 can contribute to immune evasion by influencing immune cell recruitment to the tumor, cytokine secretion in the TME, and inflammatory signaling pathways. In some contexts, p53 mutations increase neoantigen load which improves response to immune checkpoint inhibition. Therapeutic restoration of mutated p53 can restore anti-cancer immune cell infiltration and ameliorate pro-tumor signaling to induce tumor regression. Indeed, there is clinical evidence to suggest that restoring p53 can induce an anti-cancer immune response in immunologically cold tumors. Clinical trials investigating the combination of p53-restoring compounds or p53-based vaccines with immunotherapy have demonstrated anti-tumor immune activation and tumor regression with heterogeneity across cancer type. In this Review, we discuss the impact of wild-type and mutant p53 on the anti-tumor immune response, outline clinical progress as far as activating p53 to induce an immune response across a variety of cancer types, and highlight open questions limiting effective clinical translation.
Collapse
Affiliation(s)
- Lindsey Carlsen
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Legorreta Cancer Center, Brown University, Providence, RI, United States
- Pathobiology Graduate Program, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Shengliang Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Legorreta Cancer Center, Brown University, Providence, RI, United States
| | - Xiaobing Tian
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Legorreta Cancer Center, Brown University, Providence, RI, United States
| | - Arielle De La Cruz
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Legorreta Cancer Center, Brown University, Providence, RI, United States
| | - Andrew George
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Legorreta Cancer Center, Brown University, Providence, RI, United States
| | - Taylor E. Arnoff
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Legorreta Cancer Center, Brown University, Providence, RI, United States
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Legorreta Cancer Center, Brown University, Providence, RI, United States
- Pathobiology Graduate Program, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Hematology-Oncology Division, Department of Medicine, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI, United States
| |
Collapse
|
18
|
Huang G, Cierpicki T, Grembecka J. 2-Aminobenzothiazoles in anticancer drug design and discovery. Bioorg Chem 2023; 135:106477. [PMID: 36989736 PMCID: PMC10718064 DOI: 10.1016/j.bioorg.2023.106477] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/26/2023] [Accepted: 03/10/2023] [Indexed: 03/22/2023]
Abstract
Cancer is one of the major causes of mortality and morbidity worldwide. Substantial research efforts have been made to develop new chemical entities with improved anticancer efficacy. 2-Aminobenzothiazole is an important class of heterocycles containing one sulfur and two nitrogen atoms, which is associated with a broad spectrum of medical and pharmacological activities, including antitumor, antibacterial, antimalarial, anti-inflammatory, and antiviral activities. In recent years, an extraordinary collection of potent and low-toxicity 2-aminobenzothiazole compounds have been discovered as new anticancer agents. Herein, we provide a comprehensive review of this class of compounds based on their activities against tumor-related proteins, including tyrosine kinases (CSF1R, EGFR, VEGFR-2, FAK, and MET), serine/threonine kinases (Aurora, CDK, CK, RAF, and DYRK2), PI3K kinase, BCL-XL, HSP90, mutant p53 protein, DNA topoisomerase, HDAC, NSD1, LSD1, FTO, mPGES-1, SCD, hCA IX/XII, and CXCR. In addition, the anticancer potentials of 2-aminobenzothiazole-derived chelators and metal complexes are also described here. Moreover, the design strategies, mechanism of actions, structure-activity relationships (SAR) and more advanced stages of pre-clinical development of 2-aminobenzothiazoles as new anticancer agents are extensively reviewed in this article. Finally, the examples that 2-aminobenzothiazoles showcase an advantage over other heterocyclic systems are also highlighted.
Collapse
Affiliation(s)
- Guang Huang
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
19
|
Hlaváč V, Červenková L, Šůsová S, Holý P, Liška V, Vyčítal O, Šorejs O, Fiala O, Daum O, Souček P. Exome Sequencing of Paired Colorectal Carcinomas and Synchronous Liver Metastases for Prognosis and Therapy Prediction. JCO Precis Oncol 2023; 7:e2200557. [PMID: 37141551 DOI: 10.1200/po.22.00557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Abstract
PURPOSE Analysis of somatic variant profiles in retrospectively collected pairs of primary tumors and synchronous liver metastases from surgically treated patients with colorectal carcinomas. Mutational profiles were compared between groups of patients stratified by response to chemotherapy and survival. PATIENTS AND METHODS The study used whole-exome sequencing of tumor sample pairs from 20 patients diagnosed and treated at a single center. The Cancer Genome Atlas COAD-READ data set (n = 380) was used for validation in silico, where possible. RESULTS The most frequently altered oncodrivers were APC (55% in primaries and 60% in metastases), TP53 (50/45), TRIP11 (30/5), FAT4 (20/25), and KRAS (15/25). Harboring variants with a high or moderate predicted functional effect in KRAS in primary tumors was significantly associated with poor relapse-free survival in both our sample set and the validation data set. We found a number of additional prognostic associations, including mutational load, alterations in individual genes, oncodriver pathways, and single base substitution (SBS) signatures in primary tissues, which were not confirmed by validation. Altered ATM, DNAH11, and MUC5AC, or a higher share of SBS24 signature in metastases seemed to represent poor prognostic factors, but because of a lack of suitable validation data sets, these results must be treated with extreme caution. No gene or profile was significantly associated with response to chemotherapy. CONCLUSION Taken together, we report subtle differences in exome mutational profiles between paired primary tumors and synchronous liver metastases and a distinct prognostic relevance of KRAS in primary tumors. Although the general scarcity of primary tumor-synchronous metastasis sample pairs with high-quality clinical data makes robust validation difficult, this study provides potentially valuable data for utilization in precision oncology and could serve as a springboard for larger studies.
Collapse
Affiliation(s)
- Viktor Hlaváč
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Lenka Červenková
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Simona Šůsová
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Petr Holý
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Václav Liška
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Department of Surgery, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Ondřej Vyčítal
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Department of Surgery, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Ondřej Šorejs
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Department of Oncology and Radiotherapeutics, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Ondřej Fiala
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Department of Oncology and Radiotherapeutics, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Ondřej Daum
- Department of Pathology, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Souček
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| |
Collapse
|
20
|
Fallatah MMJ, Law FV, Chow WA, Kaiser P. Small-molecule correctors and stabilizers to target p53. Trends Pharmacol Sci 2023; 44:274-289. [PMID: 36964053 PMCID: PMC10511064 DOI: 10.1016/j.tips.2023.02.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/26/2023]
Abstract
The tumor suppressor p53 is the most frequently mutated protein in human cancer and tops the list of high-value precision oncology targets. p53 prevents initiation and progression of cancer by inducing cell-cycle arrest and various forms of cell death. Tumors have thus evolved ways to inactivate p53, mainly by TP53 mutations or by hyperactive p53 degradation. This review focuses on two types of p53 targeting compounds, MDM2 antagonists and mutant p53 correctors. MDM2 inhibitors prevent p53 protein degradation, while correctors restore tumor suppressor activity of p53 mutants by enhancing thermodynamic stability. Herein we explore both novel and repurposed p53 targeting compounds, discuss their mode of action, and examine the challenges in advancing them to the clinic.
Collapse
Affiliation(s)
- Maryam M J Fallatah
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Fiona V Law
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Warren A Chow
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Peter Kaiser
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
21
|
Dual roles of TRIM3 in colorectal cancer by retaining p53 in the cytoplasm to decrease its nuclear expression. Cell Death Discov 2023; 9:85. [PMID: 36894560 PMCID: PMC9998637 DOI: 10.1038/s41420-023-01386-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Colorectal cancer is a very heterogeneous disease caused by the interaction of genetic and environmental factors. P53, as a frequent mutation gene, plays a critical role in the adenoma-carcinoma transition during the tumorous pathological process. Our team discovered TRIM3 as a tumor-associated gene in CRC by high-content screening techniques. TRIM3 demonstrated both tumor-suppressive and tumorigenic features in cell experiments dependent on the cell status of wild or mutant p53. TRIM3 could directly interact with the C terminus of p53 (residues 320 to 393), a common segment of wtp53 and mutp53. Moreover, TRIM3 could exert different neoplastic features by retaining p53 in the cytoplasm to decrease its nuclear expression in a wtp53 or mutp53-dependent pathway. Chemotherapy resistance develops in nearly all patients with advanced CRC and seriously limits the therapeutic efficacies of anticancer drugs. TRIM3 could reverse the chemotherapy resistance of oxaliplatin in mutp53 CRC cells by degradation of mutp53 in the nuclei to downregulate the multidrug resistance gene. Therefore, TRIM3 could be a potential therapeutic strategy to improve the survival of CRC patients with mutp53.
Collapse
|
22
|
Luo Y. Mutation of the TP53 Gene in Placental Chorangiomatosis. Fetal Pediatr Pathol 2023; 42:400-409. [PMID: 36591909 DOI: 10.1080/15513815.2022.2161330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Objective: We explored the frequency of TP53 gene mutations in chorangiomas (CA) and chorangiomatosis (CM). Materials and -methods: By Sanger sequencing, we evaluated mutations in exons 4-6 of the TP53 gene in CM and CA regions of placentas. Results: In total, 7/11(63.6%) CAs and 24/26 (92.3%) CMs had TP53 mutations, with a significantly higher frequency in the latter. Mutations in both groups predominately involved exon 4, most commonly at the 119th C. The mutation types at the 119th C were C/G and G/G. Among the patients with exon 4 mutations at the 119th C, C/G mutations, the most common type, were observed more frequently in the CM group (63.16%, 12/19) than in the CA group (14.29%, 1/7), and the difference was significant. Conclusion: It is suggested that both CM and CA are tumors rather than tumor-like lesions. Although they are histologically similar, they have a different TP53 profile.
Collapse
Affiliation(s)
- Ying Luo
- Department of Pathology, Fujian Maternal and Child Health Hospital; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| |
Collapse
|
23
|
Zhang Z, Sun J. The Origin of Clonal Hematopoiesis and Its Implication in Human Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1442:65-83. [PMID: 38228959 DOI: 10.1007/978-981-99-7471-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Clonal expansion of hematopoietic cells is first observed in hematological malignancies where all the leukemic cells can be traced back to a single cell carrying oncogenic alterations. Interestingly, expansion of hematopoietic clones with defined genomic alterations, including single nucleotide variants (SNVs), small insertions and deletions (indels), and large structural chromosomal alterations (CAs), is also found in the healthy population. These genomic changes often affect leukemia driver genes. As a result, healthy individuals bearing such clonal hematopoiesis (CH) are at a higher risk of hematological malignancies. In addition to blood cancers, SNV/indel-related CH has been found associated with elevated cardiovascular and all-cause mortality, indicating adverse impacts of abnormalities in the blood on the normal functions of non-hematological tissues. In the past decade, much effort has been invested in understanding the origins of CH and its causal relationship with diseases in hematological and non-hematological tissues. Here, we review recent progress in these areas and discuss future directions that can be pursued to translate the acquired knowledge into better management of CH-related diseases.
Collapse
Affiliation(s)
- Zhen Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jianlong Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
24
|
Investigational Microbiological Therapy for Glioma. Cancers (Basel) 2022; 14:cancers14235977. [PMID: 36497459 PMCID: PMC9736089 DOI: 10.3390/cancers14235977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/27/2022] [Accepted: 11/29/2022] [Indexed: 12/07/2022] Open
Abstract
Glioma is the most common primary malignancy of the central nervous system (CNS), and 50% of patients present with glioblastoma (GBM), which is the most aggressive type. Currently, the most popular therapies are progressive chemotherapy and treatment with temozolomide (TMZ), but the median survival of glioma patients is still low as a result of the emergence of drug resistance, so we urgently need to find new therapies. A growing number of studies have shown that the diversity, bioactivity, and manipulability of microorganisms make microbial therapy a promising approach for cancer treatment. However, the many studies on the research progress of microorganisms and their derivatives in the development and treatment of glioma are scattered, and nobody has yet provided a comprehensive summary of them. Therefore, in this paper, we review the research progress of microorganisms and their derivatives in the development and treatment of glioma and conclude that it is possible to treat glioma by exogenous microbial therapies and targeting the gut-brain axis. In this article, we discuss the prospects and pressing issues relating to these therapies with the aim of providing new ideas for the treatment of glioma.
Collapse
|
25
|
Li F, Li S, Wang X, Liu C, Li X, Li Y, Liu Y. To investigate the prognostic factors of stage Ⅰ-Ⅱ gastric cancer based on P53 mutation and tumor budding. Pathol Res Pract 2022; 240:154195. [PMID: 36356333 DOI: 10.1016/j.prp.2022.154195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/27/2022] [Accepted: 10/29/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND P53 is a tumor suppressor and genome guardian factor, and tumor budding is a key link in tumor metastasis. The purpose of this study was to investigate P53 mutation and tumor budding in stage Ⅰ-Ⅱ gastric cancer, to explore the correlation with clinicopathological features, and to reveal the independent prognostic factors of gastric cancer. METHODS The data of 588 patients with stage Ⅰ-Ⅱ gastric cancer who underwent radical surgical resection from April 2015 to December 2016 in the Fourth Hospital of Hebei Medical University were retrospectively analyzed and followed up. Immunohistochemistry Envision method was used to conduct P53 staining for paraffin-embedded gastric cancer tissues, and ITBCC recommended tumor budding evaluation method was used to count tumor budding in gastric cancer tissues. The factors affecting the prognosis of gastric cancer were analyzed. RESULTS There were 209 cases (35.54%) of P53 wild-type and 379 cases (64.46 %) of P53 mutant in 588 patients with stage Ⅰ-Ⅱ gastric cancer. P53 mutation rate were closely correlated with Lauren classification (χ2 =8.152, p = 0.017), degree of differentiation (χ2 =10.495, p = 0.004), number of lymph node metastasis (χ2 =25.550, p < 0.001), and clinical stage (χ2 =7.617, p = 0.016). Tumor budding were closely correlated with Lauren classification (χ2 =194.533, p < 0.001), degree of tissue differentiation (χ2 =22.719, p < 0.001), depth of tumor invasion (χ2 =19.204, p = 0.004), number of lymph node metastasis (χ2 =22.555, p = 0.001), clinical stage (χ2 =10.769, p = 0.005), and vascular tumors bolt (χ2 =12.478, p = 0.002). The higher the tumor budding grade was, the higher the P53 mutation rate was (χ2 =12.933, p = 0.002). Lauren classification (p < 0.001), degree of tissue differentiation (p = 0.005), vascular tumors bolt (p < 0.001) and P53 mutation (p = 0.006) were independent influencing factors for 5-year survival of patients with stage Ⅰ-Ⅱ gastric cancer. CONCLUSION P53 mutation status is an independent prognostic factor for gastric cancer patients and a promising cancer treatment target. Tumor budding is a very reliable independent prognostic parameter with important clinical value and should be routinely reported as a biomarker.
Collapse
Affiliation(s)
- Fang Li
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shi Li
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xinran Wang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chang Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoya Li
- Department of Scientific Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yong Li
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
26
|
Mutant p53 in cancer: from molecular mechanism to therapeutic modulation. Cell Death Dis 2022; 13:974. [PMID: 36400749 PMCID: PMC9674619 DOI: 10.1038/s41419-022-05408-1] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022]
Abstract
TP53, a crucial tumor suppressor gene, is the most commonly mutated gene in human cancers. Aside from losing its tumor suppressor function, mutant p53 (mutp53) often acquires inherent, novel oncogenic functions, which is termed "gain-of-function". Emerging evidence suggests that mutp53 is highly associated with advanced malignancies and poor prognosis, which makes it a target for development of novel cancer therapies. Herein, we provide a summary of our knowledge of the mutp53 types and mutp53 spectrum in cancers. The mechanisms of mutp53 accumulation and gain-of-function are also summarized. Furthermore, we discuss the gain-of-function of mutp53 in cancers: genetic instability, ferroptosis, microenvironment, and stemness. Importantly, the role of mutp53 in the clinic is also discussed, particularly with regard to chemotherapy and radiotherapy. Last, emphasis is given to emerging strategies on how to target mutp53 for tumor therapy. Thus, this review will contribute to better understanding of the significance of mutp53 as a target for therapeutic strategies.
Collapse
|
27
|
Shi Y, Gao Q, Liu Z, Shen G, Sun X, Di X. Identification of Immune and Hypoxia Risk Classifier to Estimate Immune Microenvironment and Prognosis in Cervical Cancer. JOURNAL OF ONCOLOGY 2022; 2022:6906380. [PMID: 36304989 PMCID: PMC9593224 DOI: 10.1155/2022/6906380] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/04/2022] [Accepted: 10/01/2022] [Indexed: 11/07/2023]
Abstract
Purpose Cervical cancer (CC) is one of the most common gynecologic neoplasms. Hypoxia is an essential trigger for activating immunosuppressive activity and initiating malignant tumors. However, the determination of the role of immunity and hypoxia on the clinical outcome of CC patients remains unclear. Methods The CC independent cohort were collected from TCGA database. Consensus cluster analysis was employed to determine a molecular subtype based on immune and hypoxia gene sets. Cox relevant analyses were utilized to set up a risk classifier for prognosis assessment. The underlying pathways of classifier genes were detected by GSEA. Moreover, we conducted CIBERSORT algorithm to mirror the immune status of CC samples. Results We observed two cluster related to immune and hypoxia status and found the significant difference in outcome of patients between the two clusters. A total of 251 candidate genes were extracted from the two clusters and enrolled into Cox relevant analyses. Then, seven hub genes (CCL20, CXCL2, ITGA5, PLOD2, PTGS2, TGFBI, and VEGFA) were selected to create an immune and hypoxia-based risk classifier (IHBRC). The IHBRC can precisely distinguish patient risk and estimate clinical outcomes. In addition, IHBRC was closely bound up with tumor associated pathways such as hypoxia, P53 signaling and TGF β signaling. IHBRC was also tightly associated with numerous types of immunocytes. Conclusion This academic research revealed that IHBRC can be served as predictor for prognosis assessment and cancer treatment estimation in CC.
Collapse
Affiliation(s)
- Yujing Shi
- Department of Oncology, Jurong People's Hospital, Huayang Town, Jurong City, China
| | - Qing Gao
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zeyuan Liu
- Department of Radiation Oncology, Nanjing Jiangning Hospital and the Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Gefenqiang Shen
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinchen Sun
- Department of Oncology, Jurong People's Hospital, Huayang Town, Jurong City, China
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoke Di
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
28
|
Hlaváč V, Holý P, Václavíková R, Rob L, Hruda M, Mrhalová M, Černaj P, Bouda J, Souček P. Whole-exome sequencing of epithelial ovarian carcinomas differing in resistance to platinum therapy. Life Sci Alliance 2022; 5:5/12/e202201551. [PMID: 36229065 PMCID: PMC9574568 DOI: 10.26508/lsa.202201551] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/23/2022] [Accepted: 09/23/2022] [Indexed: 11/07/2022] Open
Abstract
Exploration of the prognostic and predictive significance of exome variation in epithelial ovarian carcinoma patients, with TP53, Hippo, homologous recombination genes, and the SBS6 signature as the most interesting results. Epithelial ovarian carcinoma (EOC) is highly fatal because of the risk of resistance to therapy and recurrence. We performed whole-exome sequencing of blood and tumor tissue pairs of 50 patients with surgically resected EOC. Compared with sensitive patients, platinum-resistant patients had a significantly higher somatic mutational rate in TP53 and lower in several genes from the Hippo pathway. We confirmed the pivotal role of somatic mutations in homologous recombination repair genes in platinum sensitivity and favorable prognosis of EOC patients. Implementing the germline homologous recombination repair profile significantly improved the prediction. In addition, distinct mutational signatures, for example, SBS6, and overall mutational load, somatic mutations in PABPC1, PABPC3, and TFAM co-segregated with the resistance status, high-grade serous carcinoma subtype, or overall survival of patients. We generated germline and somatic genetic landscapes of prognostically different subgroups of EOC patients for further follow-up studies focused on utilizing the observed associations in precision oncology.
Collapse
Affiliation(s)
- Viktor Hlaváč
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic,Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
| | - Petr Holý
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic,Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic,Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Radka Václavíková
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic,Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
| | - Lukáš Rob
- Department of Gynecology and Obstetrics, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic
| | - Martin Hruda
- Department of Gynecology and Obstetrics, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic
| | - Marcela Mrhalová
- Department of Pathology and Molecular Medicine, Second Faculty of Medicine and Motol University Hospital, Charles University, Prague, Czech Republic
| | - Petr Černaj
- Department of Gynecology and Obstetrics, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Jiří Bouda
- Department of Gynecology and Obstetrics, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Souček
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic,Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic,Correspondence:
| |
Collapse
|
29
|
Babamohamadi M, Babaei E, Ahmed Salih B, Babamohammadi M, Jalal Azeez H, Othman G. Recent findings on the role of wild-type and mutant p53 in cancer development and therapy. Front Mol Biosci 2022; 9:903075. [PMID: 36225257 PMCID: PMC9549909 DOI: 10.3389/fmolb.2022.903075] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
The p53 protein is a tumor suppressor encoded by the TP53 gene and consists of 393 amino acids with four main functional domains. This protein responds to various cellular stresses to regulate the expression of target genes, thereby causing DNA repair, cell cycle arrest, apoptosis, metabolic changes, and aging. Mutations in the TP53 gene and the functions of the wild-type p53 protein (wtp53) have been linked to various human cancers. Eight TP53 gene mutations are located in codons, constituting 28% of all p53 mutations. The p53 can be used as a biomarker for tumor progression and an excellent target for designing cancer treatment strategies. In wild-type p53-carrying cancers, abnormal signaling of the p53 pathway usually occurs due to other unusual settings, such as high MDM2 expression. These differences between cancer cell p53 and normal cells have made p53 one of the most important targets for cancer treatment. In this review, we have dealt with various issues, such as the relative contribution of wild-type p53 loss of function, including transactivation-dependent and transactivation-independent activities in oncogenic processes and their role in cancer development. We also discuss the role of p53 in the process of ferroptosis and its targeting in cancer treatment. Finally, we focus on p53-related drug delivery systems and investigate the challenges and solutions.
Collapse
Affiliation(s)
- Mehregan Babamohamadi
- Department of Biology, School of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Esmaeil Babaei
- Department of Biology, School of Natural Sciences, University of Tabriz, Tabriz, Iran
- Interfaculty Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Tübingen, Germany
- *Correspondence: Esmaeil Babaei,
| | - Burhan Ahmed Salih
- Department of Medical Laboratory Technology, Erbil Health and Medical Technical College, Erbil Polytechnic University, Erbil, Iraq
- Department of Medical Laboratory Technology, AlQalam University College, Kirkuk, Iraq
| | - Mahshid Babamohammadi
- Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hewa Jalal Azeez
- Department of Biology, School of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Goran Othman
- Department of Medical Laboratory Technology, Erbil Health and Medical Technical College, Erbil Polytechnic University, Erbil, Iraq
- Department of Medical Laboratory Technology, AlQalam University College, Kirkuk, Iraq
| |
Collapse
|