1
|
Haripriya S, Vijayalakshmi M, Ala C, Murugesan S, Pavadai P, Kunjiappan S, Pandian SRK. Pharmacoinformatics-based prediction of Checkpoint kinase-1 inhibitors from Momordica charantia Linn. for cancer. Comput Biol Chem 2025; 115:108286. [PMID: 39612740 DOI: 10.1016/j.compbiolchem.2024.108286] [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: 06/14/2024] [Revised: 10/25/2024] [Accepted: 11/17/2024] [Indexed: 12/01/2024]
Abstract
Checkpoint kinase 1 (Chk-1), a serine/threonine kinase family protein, is an emerging target in cancer research owing to its crucial role in cell cycle arrest. Therefore, we aimed to predict potential Chk-1 inhibitors from Momordica charantia Linn., using high-throughput molecular docking. We used a graph theoretical network approach to determine the target protein, Chk-1. Among 86 compounds identified from M. charantia L., five molecules such as α-spinasterol (-9.7 kcal × mol-1), stigmasterol (-9.6 kcal × mol-1), stigmasta-7,22,25-trienol (-9.5 kcal × mol-1), campesterol (-9.5 kcal × mol-1), and stigmasta-7,25-dien-3beta-ol (-9.5 kcal × mol-1) and standard drug CCT245737 (-8.3 kcal × mol-1) displayed highest binding affinity with Chk-1. Besides, pharmacokinetic studies have demonstrated the non-toxic and drug-like properties of these compounds. Furthermore, molecular dynamics (MD) simulation studies confirmed the strong intermolecular interactions and stability of the compounds with Chk-1. The estimation of binding free-energy derived from molecular docking was fully recognized by the Molecular Mechanics-Generalized Born Surface Area (MM-GBSA) produced from the MD simulation paths. Altogether, these five compounds may serve as effective inhibitors of Chk-1, thereby could be used to develop new medications for cancer treatment.
Collapse
Affiliation(s)
- Subramanian Haripriya
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, Tamil Nadu 626126, India
| | - Muniyandi Vijayalakshmi
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, Tamil Nadu 626126, India
| | - Chandu Ala
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Rajasthan 333031, India
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Rajasthan 333031, India
| | - Parasuraman Pavadai
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bengaluru, Karnataka 560054, India
| | - Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, Tamil Nadu 626126, India
| | - Sureshbabu Ram Kumar Pandian
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, Tamil Nadu 626126, India.
| |
Collapse
|
2
|
Liu Y, Wang Y, Tan S, Shi X, Wen J, Chen D, Zhao Y, Pan W, Jia Z, Lu C, Lou G. Characterization of G2/M checkpoint classifier for personalized treatment in uterine corpus endometrial carcinoma. Cancer Cell Int 2025; 25:34. [PMID: 39920729 PMCID: PMC11806828 DOI: 10.1186/s12935-025-03667-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Uterine Corpus Endometrial Carcinoma (UCEC) is a highly heterogeneous tumor, and limitations in current diagnostic methods, along with treatment resistance in some patients, pose significant challenges for managing UCEC. The excessive activation of G2/M checkpoint genes is a crucial factor affecting malignancy prognosis and promoting treatment resistance. METHODS Gene expression profiles and clinical feature data mainly came from the TCGA-UCEC cohort. Unsupervised clustering was performed to construct G2/M checkpoint (G2MC) subtypes. The differences in biological and clinical features of different subtypes were compared through survival analysis, clinical characteristics, immune infiltration, tumor mutation burden, and drug sensitivity analysis. Ultimately, an artificial neural network (ANN) and machine learning were employed to develop the G2MC subtypes classifier. RESULTS We constructed a classifier based on the overall activity of the G2/M checkpoint signaling pathway to identify patients with different risks and treatment responses, and attempted to explore potential therapeutic targets. The results showed that two G2MC subtypes have completely different G2/M checkpoint-related gene expression profiles. Compared with the subtype C2, the subtype C1 exhibited higher G2MC scores and was associated with faster disease progression, higher clinical staging, poorer pathological types, and lower therapy responsiveness of cisplatin, radiotherapy and immunotherapy. Experiments targeting the feature gene KIF23 revealed its crucial role in reducing HEC-1A sensitivity to cisplatin and radiotherapy. CONCLUSION In summary, our study developed a classifier for identifying G2MC subtypes, and this finding holds promise for advancing precision treatment strategies for UCEC.
Collapse
Affiliation(s)
- Yiming Liu
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yusi Wang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Shu Tan
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaochen Shi
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jinglin Wen
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Dejia Chen
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yue Zhao
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wenjing Pan
- Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhaoyang Jia
- Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chunru Lu
- Department of Gynecology, Suihua Maternity and Health Care Hospital, Suihua, China.
| | - Ge Lou
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
3
|
Coquel F, Ho SZ, Tsai KC, Yang CY, Aze A, Devin J, Chang TH, Kong-Hap M, Bioteau A, Moreaux J, Maiorano D, Pourquier P, Yang WC, Lin YL, Pasero P. Synergistic effect of inhibiting CHK2 and DNA replication on cancer cell growth. eLife 2025; 13:RP104718. [PMID: 39887032 PMCID: PMC11785374 DOI: 10.7554/elife.104718] [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: 02/01/2025] Open
Abstract
Cancer cells display high levels of oncogene-induced replication stress (RS) and rely on DNA damage checkpoint for viability. This feature is exploited by cancer therapies to either increase RS to unbearable levels or inhibit checkpoint kinases involved in the DNA damage response. Thus far, treatments that combine these two strategies have shown promise but also have severe adverse effects. To identify novel, better-tolerated anticancer combinations, we screened a collection of plant extracts and found two natural compounds from the plant, Psoralea corylifolia, that synergistically inhibit cancer cell proliferation. Bakuchiol inhibited DNA replication and activated the checkpoint kinase CHK1 by targeting DNA polymerases. Isobavachalcone interfered with DNA double-strand break repair by inhibiting the checkpoint kinase CHK2 and DNA end resection. The combination of bakuchiol and isobavachalcone synergistically inhibited cancer cell proliferation in vitro. Importantly, it also prevented tumor development in xenografted NOD/SCID mice. The synergistic effect of inhibiting DNA replication and CHK2 signaling identifies a vulnerability of cancer cells that might be exploited by using clinically approved inhibitors in novel combination therapies.
Collapse
Affiliation(s)
- Flavie Coquel
- Institut de Génétique Humaine, Univ. de Montpellier, CNRSMontpellierFrance
- ‘Maintenance of Genome Integrity during DNA replication’ laboratory, équipe labélisée Ligue contre le CancerMontpellierFrance
| | - Sing-Zong Ho
- Agricultural Biotechnology Research Center, Academia SinicaTaipeiTaiwan
| | - Keng-Chang Tsai
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical UniversityTaipeiTaiwan
- National Research Institute of Chinese Medicine, Ministry of Health and WelfareTaipeiTaiwan
| | - Chun-Yen Yang
- Institut de Génétique Humaine, Univ. de Montpellier, CNRSMontpellierFrance
| | - Antoine Aze
- Institut de Génétique Humaine, Univ. de Montpellier, CNRSMontpellierFrance
- ‘Genome Surveillance and Stability’ Laboratory, IGH, Univ. de Montpellier, CNRSMontpellierFrance
| | - Julie Devin
- Institut de Génétique Humaine, Univ. de Montpellier, CNRSMontpellierFrance
- ‘Normal and Malignant B cells’ laboratory', IGH, Univ. de Montpellier, CNRSMontpellierFrance
| | - Ting-Hsiang Chang
- Agricultural Biotechnology Research Center, Academia SinicaTaipeiTaiwan
| | - Marie Kong-Hap
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut régional du Cancer de MontpellierMontpellierFrance
| | - Audrey Bioteau
- Institut de Génétique Humaine, Univ. de Montpellier, CNRSMontpellierFrance
- ‘Maintenance of Genome Integrity during DNA replication’ laboratory, équipe labélisée Ligue contre le CancerMontpellierFrance
| | - Jerome Moreaux
- Institut de Génétique Humaine, Univ. de Montpellier, CNRSMontpellierFrance
- ‘Normal and Malignant B cells’ laboratory', IGH, Univ. de Montpellier, CNRSMontpellierFrance
- Institut Universitaire de FranceParisFrance
- Department of Biological Hematology, CHU MontpellierMontpellierFrance
| | - Domenico Maiorano
- Institut de Génétique Humaine, Univ. de Montpellier, CNRSMontpellierFrance
- ‘Genome Surveillance and Stability’ Laboratory, IGH, Univ. de Montpellier, CNRSMontpellierFrance
| | - Philippe Pourquier
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut régional du Cancer de MontpellierMontpellierFrance
| | - Wen-Chin Yang
- Agricultural Biotechnology Research Center, Academia SinicaTaipeiTaiwan
- Graduate Institute of Integrated Medicine, China Medical UniversityTaichungTaiwan
- Department of Life Sciences, National Chung-Hsing UniversityTaichungTaiwan
| | - Yea-Lih Lin
- ‘Maintenance of Genome Integrity during DNA replication’ laboratory, équipe labélisée Ligue contre le CancerMontpellierFrance
| | - Philippe Pasero
- Institut de Génétique Humaine, Univ. de Montpellier, CNRSMontpellierFrance
- ‘Maintenance of Genome Integrity during DNA replication’ laboratory, équipe labélisée Ligue contre le CancerMontpellierFrance
| |
Collapse
|
4
|
Lee JM, Lee WH, Cho SH, Park JW, Kwon HN, Kim JH, Lee SH, Yoon JH, Park S, Kim SC. DRG2 levels in prostate cancer cell lines predict response to PARP inhibitor during docetaxel treatment. Investig Clin Urol 2025; 66:56-66. [PMID: 39791585 PMCID: PMC11729229 DOI: 10.4111/icu.20240263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 01/30/2025] Open
Abstract
PURPOSE Developmentally regulated GTP-binding protein 2 (DRG2) regulates microtubule dynamics and G2/M arrest during docetaxel treatment. Poly ADP-ribose polymerase (PARP) acts as an important repair system for DNA damage caused by docetaxel treatment. This study investigated whether DRG2 expression affects response to PARP inhibitors (olaparib) using prostate cancer cell lines PC3, DU145, LNCaP-FGC, and LNCaP-LN3. MATERIALS AND METHODS The cell viability and DRG2 expression levels were assessed using colorimetric-based cell viability assay and western blot. Cells were transfected with DRG2 siRNA, and pcDNA6/V5-DRG2 was used to overexpress DRG2. Flow cytometry was applied for cell cycle assay and apoptosis analysis using the Annexing V cell death assay. RESULTS The expression of DRG2 was highest in LNCaP-LN3 and lowest in DU145 cells. Expressions of p53 in PC3, DU145, and the two LNCaP cell lines were null-type, high-expression, and medium-expression, respectively. In PC3 (DRG2 high, p53 null) cells, docetaxel increased G2/M arrest without apoptosis; however, subsequent treatment with olaparib promoted apoptosis. In DU145 and LNCaP-FGC (DRG2 low), docetaxel increased sub-G1 but not G2/M arrest and induced apoptosis, whereas olaparib had no additional effect. In LNCaP-LN3 (DRG2 high, p53 wild-type), docetaxel increased sub-G1 and G2/M arrest, furthermore olaparib enhanced cell death. Docetaxel and olaparib combination treatment had a slight effect on DRG2 knockdown PC3, but increased apoptosis in DRG2-overexpressed DU145 cells. CONCLUSIONS DRG2 and p53 expressions play an important role in prostate cancer cell lines treated with docetaxel, and DRG2 levels can predict the response to PARP inhibitors.
Collapse
Affiliation(s)
- Jeong Min Lee
- School of Biological Sciences, University of Ulsan, Ulsan, Korea
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
| | - Won Hyeok Lee
- School of Biological Sciences, University of Ulsan, Ulsan, Korea
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
| | - Seung Hyeon Cho
- School of Biological Sciences, University of Ulsan, Ulsan, Korea
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
| | - Jeong Woo Park
- School of Biological Sciences, University of Ulsan, Ulsan, Korea
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
| | - Hyuk Nam Kwon
- School of Biological Sciences, University of Ulsan, Ulsan, Korea
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
| | - Ji Hye Kim
- Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Sang Hun Lee
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
- Department of Obstetrics & Gynecology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Ji Hyung Yoon
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
- Department of Urology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Sungchan Park
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
- Department of Urology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Seong Cheol Kim
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
- Department of Urology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea.
| |
Collapse
|
5
|
Joshi JN, Lerner AD, Scallo F, Grumet AN, Matteson P, Millonig JH, Valvezan AJ. mTORC1 activity oscillates throughout the cell cycle, promoting mitotic entry and differentially influencing autophagy induction. Cell Rep 2024; 43:114543. [PMID: 39067023 DOI: 10.1016/j.celrep.2024.114543] [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] [Revised: 06/07/2024] [Accepted: 07/09/2024] [Indexed: 07/30/2024] Open
Abstract
Mechanistic Target of Rapamycin Complex 1 (mTORC1) is a master metabolic regulator that is active in nearly all proliferating eukaryotic cells; however, it is unclear whether mTORC1 activity changes throughout the cell cycle. We find that mTORC1 activity oscillates from lowest in mitosis/G1 to highest in S/G2. The interphase oscillation is mediated through the TSC complex but is independent of major known regulatory inputs, including Akt and Mek/Erk signaling. By contrast, suppression of mTORC1 activity in mitosis does not require the TSC complex. mTORC1 has long been known to promote progression through G1. We find that mTORC1 also promotes progression through S and G2 and is important for satisfying the Chk1/Wee1-dependent G2/M checkpoint to allow entry into mitosis. We also find that low mTORC1 activity in G1 sensitizes cells to autophagy induction in response to partial mTORC1 inhibition or reduced nutrient levels. Together, these findings demonstrate that mTORC1 is differentially regulated throughout the cell cycle, with important phase-specific consequences for proliferating cells.
Collapse
Affiliation(s)
- Jay N Joshi
- Molecular Biosciences Program, Rutgers University, Piscataway, NJ, USA; Center for Advanced Biotechnology and Medicine, Piscataway, NJ, USA
| | - Ariel D Lerner
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ, USA
| | - Frank Scallo
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ, USA
| | | | - Paul Matteson
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ, USA
| | - James H Millonig
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ, USA; Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Alexander J Valvezan
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ, USA; Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
6
|
Zhao Y, Short NJ, Kantarjian HM, Chang TC, Ghate PS, Qu C, Macaron W, Jain N, Thakral B, Phillips AH, Khoury J, Garcia-Manero G, Zhang W, Fan Y, Yang H, Garris RS, Nasr LF, Kriwacki RW, Roberts KG, Konopleva M, Jabbour EJ, Mullighan CG. Genomic determinants of response and resistance to inotuzumab ozogamicin in B-cell ALL. Blood 2024; 144:61-73. [PMID: 38551807 PMCID: PMC11251222 DOI: 10.1182/blood.2024023930] [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: 01/17/2024] [Revised: 02/28/2024] [Accepted: 03/15/2024] [Indexed: 04/09/2024] Open
Abstract
ABSTRACT Inotuzumab ozogamicin (InO) is an antibody-drug conjugate that delivers calicheamicin to CD22-expressing cells. In a retrospective cohort of InO-treated patients with B-cell acute lymphoblastic leukemia, we sought to understand the genomic determinants of the response and resistance to InO. Pre- and post-InO-treated patient samples were analyzed by whole genome, exome, and/or transcriptome sequencing. Acquired CD22 mutations were observed in 11% (3/27) of post-InO-relapsed tumor samples, but not in refractory samples (0/16). There were multiple CD22 mutations per sample and the mechanisms of CD22 escape included epitope loss (protein truncation and destabilization) and epitope alteration. Two CD22 mutant cases were post-InO hyper-mutators resulting from error-prone DNA damage repair (nonhomologous/alternative end-joining repair, or mismatch repair deficiency), suggesting that hypermutation drove escape from CD22-directed therapy. CD22-mutant relapses occurred after InO and subsequent hematopoietic stem cell transplantation (HSCT), suggesting that InO eliminated the predominant clones, leaving subclones with acquired CD22 mutations that conferred resistance to InO and subsequently expanded. Acquired loss-of-function mutations in TP53, ATM, and CDKN2A were observed, consistent with a compromise of the G1/S DNA damage checkpoint as a mechanism for evading InO-induced apoptosis. Genome-wide CRISPR/Cas9 screening of cell lines identified DNTT (terminal deoxynucleotidyl transferase) loss as a marker of InO resistance. In conclusion, genetic alterations modulating CD22 expression and DNA damage response influence InO efficacy. Our findings highlight the importance of defining the basis of CD22 escape and eradication of residual disease before HSCT. The identified mechanisms of escape from CD22-targeted therapy extend beyond antigen loss and provide opportunities to improve therapeutic approaches and overcome resistance. These trials were registered at www.ClinicalTrials.gov as NCT01134575, NCT01371630, and NCT03441061.
Collapse
Affiliation(s)
- Yaqi Zhao
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
- Center of Excellence for Leukemia Studies, St. Jude Children’s Research Hospital, Memphis, TN
| | | | | | - Ti-Cheng Chang
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Pankaj S. Ghate
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
- Center of Excellence for Leukemia Studies, St. Jude Children’s Research Hospital, Memphis, TN
| | - Chunxu Qu
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
- Center of Excellence for Leukemia Studies, St. Jude Children’s Research Hospital, Memphis, TN
| | - Walid Macaron
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX
| | - Nitin Jain
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX
| | - Beenu Thakral
- Department of Hematopathology, MD Anderson Cancer Center, Houston, TX
| | - Aaron H. Phillips
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Joseph Khoury
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | | | - Wenchao Zhang
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Yiping Fan
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Hui Yang
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX
| | | | - Lewis F. Nasr
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX
| | - Richard W. Kriwacki
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Kathryn G. Roberts
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
- Center of Excellence for Leukemia Studies, St. Jude Children’s Research Hospital, Memphis, TN
| | - Marina Konopleva
- Department of Oncology and Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY
| | | | - Charles G. Mullighan
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
- Center of Excellence for Leukemia Studies, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
7
|
Li J, Cheng C, Zhang J. An analysis of AURKB's prognostic and immunological roles across various cancers. J Cell Mol Med 2024; 28:e18475. [PMID: 38898693 PMCID: PMC11187167 DOI: 10.1111/jcmm.18475] [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: 05/14/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Aurora kinase B (AURKB), an essential regulator in the process of mitosis, has been revealed through various studies to have a significant role in cancer development and progression. However, the specific mechanisms remain poorly understood. This study, therefore, seeks to elucidate the multifaceted role of AURKB in diverse cancer types. This study utilized bioinformatics techniques to examine the transcript, protein, promoter methylation and mutation levels of AURKB. The study further analysed associations between AURKB and factors such as prognosis, pathological stage, biological function, immune infiltration, tumour mutational burden (TMB) and microsatellite instability (MSI). In addition, immunohistochemical staining data of 50 cases of renal clear cell carcinoma and its adjacent normal tissues were collected to verify the difference in protein expression of AURKB in the two tissues. The results show that AURKB is highly expressed in most cancers, and the protein level of AURKB and the methylation level of its promoter vary among cancer types. Survival analysis showed that AURKB was associated with overall survival in 12 cancer types and progression-free survival in 11 cancer types. Elevated levels of AURKB were detected in the advanced stages of 10 different cancers. AURKB has a potential impact on cancer progression through its effects on cell cycle regulation as well as inflammatory and immune-related pathways. We observed a strong association between AURKB and immune cell infiltration, immunomodulatory factors, TMB and MSI. Importantly, we confirmed that the AURKB protein is highly expressed in kidney renal clear cell carcinoma (KIRC). Our study reveals that AURKB may be a potential biomarker for pan-cancer and KIRC.
Collapse
Affiliation(s)
- Jun Li
- Department of UrologyThe First Affiliated Hospital of Bengbu Medical UniversityBengbuChina
| | - Cui Cheng
- Department of Gynaecological OncologyThe First Affiliated Hospital of Bengbu Medical UniversityBengbuChina
| | - Jiajun Zhang
- Department of UrologyThe First Affiliated Hospital of Bengbu Medical UniversityBengbuChina
| |
Collapse
|
8
|
Wang Z, Zheng Z, Wang B, Zhan C, Yuan X, Lin X, Xin Q, Zhong Z, Qiu X. Characterization of a G2M checkpoint-related gene model and subtypes associated with immunotherapy response for clear cell renal cell carcinoma. Heliyon 2024; 10:e29289. [PMID: 38617927 PMCID: PMC11015143 DOI: 10.1016/j.heliyon.2024.e29289] [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: 02/06/2024] [Revised: 03/28/2024] [Accepted: 04/04/2024] [Indexed: 04/16/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) presents challenges in early diagnosis and effective treatment. In this study, we aimed to establish a prognostic model based on G2M checkpoint-related genes and identify associated clusters in ccRCC through clinical bioinformatic analysis and experimental validation. Utilizing a single-cell RNA dataset (GSE159115) and bulk-sequencing data from The Cancer Genome Atlas (TCGA) database, we analyzed the G2M checkpoint pathway in ccRCC. Differential expression analysis identified 45 genes associated with the G2M checkpoint, leading to the construction of a predictive model with four key genes (E2F2, GTSE1, RAD54L, and UBE2C). The model demonstrated reliable predictive ability for 1-, 3-, and 5-year overall survival, with AUC values of 0.794, 0.790, and 0.794, respectively. Patients in the high-risk group exhibited a worse prognosis, accompanied by significant differences in immune cell infiltration, immune function, TIDE and IPS scores, and drug sensitivities. Two clusters of ccRCC were identified using the "ConsensusClusterPlus" package, cluster 1 exhibited a worse survival rate and was resistant to chemotherapeutic drugs of Axitinib, Erlotinib, Pazopanib, Sunitinib, and Temsirolimus, but not Sorafenib. Targeted experiments on RAD54L, a gene involved in DNA repair processes, revealed its crucial role in inhibiting proliferation, invasion, and migration in 786-O cells. In conclusion, our study offers valuable insights into the molecular mechanisms underlying ccRCC, identifying potential prognostic genes and molecular subtypes associated with the G2M checkpoint. These findings hold promise for guiding personalized treatment strategies in the management of ccRCC.
Collapse
Affiliation(s)
- Zhenwei Wang
- Department of Urology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Zongtai Zheng
- Department of Urology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Bangqi Wang
- Department of Urology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Changxin Zhan
- Department of Urology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Xuefeng Yuan
- Department of Urology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoqi Lin
- Department of Urology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Qifan Xin
- Department of Urology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Zhihui Zhong
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, 525200, Guangdong, China
| | - Xiaofu Qiu
- Department of Urology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
9
|
Joshi JN, Lerner AD, Scallo F, Grumet AN, Matteson P, Millonig JH, Valvezan AJ. mTORC1 activity oscillates throughout the cell cycle promoting mitotic entry and differentially influencing autophagy induction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.06.579216. [PMID: 38370755 PMCID: PMC10871213 DOI: 10.1101/2024.02.06.579216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Mechanistic Target of Rapamycin Complex 1 (mTORC1) is a master metabolic regulator that stimulates anabolic cell growth while suppressing catabolic processes such as autophagy. mTORC1 is active in most, if not all, proliferating eukaryotic cells. However, it remains unclear whether and how mTORC1 activity changes from one cell cycle phase to another. Here we tracked mTORC1 activity through the complete cell cycle and uncover oscillations in its activity. We find that mTORC1 activity peaks in S and G2, and is lowest in mitosis and G1. We further demonstrate that multiple mechanisms are involved in controlling this oscillation. The interphase oscillation is mediated through the TSC complex, an upstream negative regulator of mTORC1, but is independent of major known regulatory inputs to the TSC complex, including Akt, Mek/Erk, and CDK4/6 signaling. By contrast, suppression of mTORC1 activity in mitosis does not require the TSC complex, and instead involves CDK1-dependent control of the subcellular localization of mTORC1 itself. Functionally, we find that in addition to its well-established role in promoting progression through G1, mTORC1 also promotes progression through S and G2, and is important for satisfying the Wee1- and Chk1- dependent G2/M checkpoint to allow entry into mitosis. We also find that low mTORC1 activity in G1 sensitizes cells to autophagy induction in response to partial mTORC1 inhibition or reduced nutrient levels. Together these findings demonstrate that mTORC1 is differentially regulated throughout the cell cycle, with important phase-specific functional consequences in proliferating cells.
Collapse
Affiliation(s)
- Jay N. Joshi
- Molecular Biosciences Program, Rutgers University, Piscataway, NJ, USA
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ, USA
| | - Ariel D. Lerner
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ, USA
| | - Frank Scallo
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ, USA
- Present affiliation: Yale School of Medicine, New Haven, CT, USA
| | | | - Paul Matteson
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ, USA
| | - James H. Millonig
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ, USA
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Alexander J. Valvezan
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ, USA
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
10
|
Leem J, Lee C, Choi DY, Oh JS. Distinct characteristics of the DNA damage response in mammalian oocytes. Exp Mol Med 2024; 56:319-328. [PMID: 38355825 PMCID: PMC10907590 DOI: 10.1038/s12276-024-01178-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/15/2023] [Accepted: 12/07/2023] [Indexed: 02/16/2024] Open
Abstract
DNA damage is a critical threat that poses significant challenges to all cells. To address this issue, cells have evolved a sophisticated molecular and cellular process known as the DNA damage response (DDR). Among the various cell types, mammalian oocytes, which remain dormant in the ovary for extended periods, are particularly susceptible to DNA damage. The occurrence of DNA damage in oocytes can result in genetic abnormalities, potentially leading to infertility, birth defects, and even abortion. Therefore, understanding how oocytes detect and repair DNA damage is of paramount importance in maintaining oocyte quality and preserving fertility. Although the fundamental concept of the DDR is conserved across various cell types, an emerging body of evidence reveals striking distinctions in the DDR between mammalian oocytes and somatic cells. In this review, we highlight the distinctive characteristics of the DDR in oocytes and discuss the clinical implications of DNA damage in oocytes.
Collapse
Affiliation(s)
- Jiyeon Leem
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Crystal Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Da Yi Choi
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Jeong Su Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea.
| |
Collapse
|
11
|
Guo H, Yu R, Zhang H, Wang W. Cytokine, chemokine alterations and immune cell infiltration in Radiation-induced lung injury: Implications for prevention and management. Int Immunopharmacol 2024; 126:111263. [PMID: 38000232 DOI: 10.1016/j.intimp.2023.111263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/11/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023]
Abstract
Radiation therapy is one of the primary treatments for thoracic malignancies, with radiation-induced lung injury (RILI) emerging as its most prevalent complication. RILI encompasses early-stage radiation pneumonitis (RP) and the subsequent development of radiation pulmonary fibrosis (RPF). During radiation treatment, not only are tumor cells targeted, but normal tissue cells, including alveolar epithelial cells and vascular endothelial cells, also sustain damage. Within the lungs, ionizing radiation boosts the intracellular levels of reactive oxygen species across various cell types. This elevation precipitates the release of cytokines and chemokines, coupled with the infiltration of inflammatory cells, culminating in the onset of RP. This pulmonary inflammatory response can persist, spanning a duration from several months to years, ultimately progressing to RPF. This review aims to explore the alterations in cytokine and chemokine release and the influx of immune cells post-ionizing radiation exposure in the lungs, offering insights for the prevention and management of RILI.
Collapse
Affiliation(s)
- Haochun Guo
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Ran Yu
- Department of Radiotherapy, Lianshui People's Hospital, Kangda College of Nanjing Medical University, Huai'an 223400, China; Jiangsu Nursing Vocational and Technical College, Huai'an 223400, China; School of Clinical Medicine, Medical College of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou 225009, China
| | - Haijun Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China.
| | - Wanpeng Wang
- Department of Radiotherapy, Lianshui People's Hospital, Kangda College of Nanjing Medical University, Huai'an 223400, China; Jiangsu Nursing Vocational and Technical College, Huai'an 223400, China; School of Clinical Medicine, Medical College of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou 225009, China.
| |
Collapse
|
12
|
Fernández-Lázaro D, Sanz B, Seco-Calvo J. The Mechanisms of Regulated Cell Death: Structural and Functional Proteomic Pathways Induced or Inhibited by a Specific Protein-A Narrative Review. Proteomes 2024; 12:3. [PMID: 38250814 PMCID: PMC10801515 DOI: 10.3390/proteomes12010003] [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: 12/04/2023] [Revised: 12/30/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
Billions of cells die in us every hour, and our tissues do not shrink because there is a natural regulation where Cell Death (CD) is balanced with cell division. The process in which cells eliminate themselves in a controlled manner is called Programmed Cell Death (PCD). The PCD plays an important role during embryonic development, in maintaining homeostasis of the body's tissues, and in the elimination of damaged cells, under a wide range of physiological and developmental stimuli. A multitude of protein mediators of PCD have been identified and signals have been found to utilize common pathways elucidating the proteins involved. This narrative review focuses on caspase-dependent and caspase-independent PCD pathways. Included are studies of caspase-dependent PCD such as Anoikis, Catastrophe Mitotic, Pyroptosis, Emperitosis, Parthanatos and Cornification, and Caspase-Independent PCD as Wallerian Degeneration, Ferroptosis, Paraptosis, Entosis, Methuosis, and Extracellular Trap Abnormal Condition (ETosis), as well as neutrophil extracellular trap abnormal condition (NETosis) and Eosinophil Extracellular Trap Abnormal Condition (EETosis). Understanding PCD from those reported in this review could shed substantial light on the processes of biological homeostasis. In addition, identifying specific proteins involved in these processes is mandatory to identify molecular biomarkers, as well as therapeutic targets. This knowledge could provide the ability to modulate the PCD response and could lead to new therapeutic interventions in a wide range of diseases.
Collapse
Affiliation(s)
- Diego Fernández-Lázaro
- Department of Cellular Biology, Genetics, Histology and Pharmacology, Faculty of Health Sciences, University of Valladolid, Campus of Soria, 42004 Soria, Spain
- Neurobiology Research Group, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain
- SARCELLOMICS Research Group, 27071 León, Spain; (B.S.); (J.S.-C.)
| | - Begoña Sanz
- SARCELLOMICS Research Group, 27071 León, Spain; (B.S.); (J.S.-C.)
- Department of Physiology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Jesús Seco-Calvo
- SARCELLOMICS Research Group, 27071 León, Spain; (B.S.); (J.S.-C.)
- Department of Physiology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Institute of Biomedicine (IBIOMED), Universidad de León, 27071 León, Spain
| |
Collapse
|
13
|
Orozco-Moreno M, Visser EA, Hodgson K, Hipgrave Ederveen AL, Bastian K, Goode EA, Öztürk Ö, Pijnenborg JFA, Eerden N, Moons SJ, Rossing E, Wang N, de Haan N, Büll C, Boltje TJ, Munkley J. Targeting aberrant sialylation and fucosylation in prostate cancer cells using potent metabolic inhibitors. Glycobiology 2023; 33:1155-1171. [PMID: 37847613 PMCID: PMC10876042 DOI: 10.1093/glycob/cwad085] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 10/09/2023] [Accepted: 10/14/2023] [Indexed: 10/19/2023] Open
Abstract
Aberrant glycosylation is a hallmark of cancer and is not just a consequence, but also a driver of a malignant phenotype. In prostate cancer, changes in fucosylated and sialylated glycans are common and this has important implications for tumor progression, metastasis, and immune evasion. Glycans hold huge translational potential and new therapies targeting tumor-associated glycans are currently being tested in clinical trials for several tumor types. Inhibitors targeting fucosylation and sialylation have been developed and show promise for cancer treatment, but translational development is hampered by safety issues related to systemic adverse effects. Recently, potent metabolic inhibitors of sialylation and fucosylation were designed that reach higher effective concentrations within the cell, thereby rendering them useful tools to study sialylation and fucosylation as potential candidates for therapeutic testing. Here, we investigated the effects of global metabolic inhibitors of fucosylation and sialylation in the context of prostate cancer progression. We find that these inhibitors effectively shut down the synthesis of sialylated and fucosylated glycans to remodel the prostate cancer glycome with only minor apparent side effects on other glycan types. Our results demonstrate that treatment with inhibitors targeting fucosylation or sialylation decreases prostate cancer cell growth and downregulates the expression of genes and proteins important in the trajectory of disease progression. We anticipate our findings will lead to the broader use of metabolic inhibitors to explore the role of fucosylated and sialylated glycans in prostate tumor pathology and may pave the way for the development of new therapies for prostate cancer.
Collapse
Affiliation(s)
- Margarita Orozco-Moreno
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Central Parkway, Newcastle-upon-Tyne, Tyne and Wear NE1 3BZ, United Kingdom
| | - Eline A Visser
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | - Kirsty Hodgson
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Central Parkway, Newcastle-upon-Tyne, Tyne and Wear NE1 3BZ, United Kingdom
| | - Agnes L Hipgrave Ederveen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Kayla Bastian
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Central Parkway, Newcastle-upon-Tyne, Tyne and Wear NE1 3BZ, United Kingdom
| | - Emily Archer Goode
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Central Parkway, Newcastle-upon-Tyne, Tyne and Wear NE1 3BZ, United Kingdom
| | - Özden Öztürk
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | | | - Nienke Eerden
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
- GlycoTherapeutics B.V., Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | - Sam J Moons
- Synvenio B.V., Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | - Emiel Rossing
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | - Ning Wang
- The Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, The University of Sheffield, Medical School, Beech Hill Rd, Sheffield, Yorkshire S10 2RX, United Kingdom
| | - Noortje de Haan
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Christian Büll
- Biomolecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Thomas J Boltje
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | - Jennifer Munkley
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Central Parkway, Newcastle-upon-Tyne, Tyne and Wear NE1 3BZ, United Kingdom
| |
Collapse
|
14
|
Duabil AJN, Cooper CR, Aldujaily E, Halford SER, Hirschberg S, Katugampola SD, Jones GDD. Investigations of the novel checkpoint kinase 1 inhibitor SRA737 in non-small cell lung cancer and colorectal cancer cells of differing tumour protein 53 gene status. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:1210-1226. [PMID: 38214010 PMCID: PMC10776598 DOI: 10.37349/etat.2023.00193] [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: 06/10/2023] [Accepted: 10/16/2023] [Indexed: 01/13/2024] Open
Abstract
Aim In response to DNA damage the serine/threonine-specific protein kinase checkpoint kinase 1 (CHK1) is activated allowing cells to enter S phase (S) and G2 phase (G2) cell-cycle arrest. CHK1 inhibitors are expected to prevent cells from entering such arrest, thereby enhancing DNA damage-induced cytotoxicity. In contrast, normal cells with intact ataxia-telangiectasia mutated (ATM), CHK2 and tumour suppressor protein 53 (P53) signalling are still able to enter cell-cycle arrest using the functioning G1/S checkpoint, thereby being rescued from enhanced cytotoxicity. The main objective of this work is to investigate the in vitro effects of the novel CHK1 inhibitor SRA737 on pairs of non-small cell lung cancer (NSCLC) and colorectal cancer (CRC) cell lines, all with genetic aberrations rendering them susceptible to replication stress but of differing tumour protein 53 (TP53) gene status, focusing on DNA damage induction and the subsequent effects on cell proliferation and viability. Methods NSCLC cell lines H23 [TP53 mutant (MUT)] and A549 [TP53 wild-type (WT)] and CRC cell lines HT29 (TP53 MUT) and HCT116 (TP53 WT) were incubated with differing micromolar concentrations of SRA737 for 24 h and then analysed using alkaline comet and phosphorylated H2A.X variant histone (γH2AX)-foci assays to assess mostly DNA single strand break and double strand break damage, respectively. Cell-counting/trypan blue staining was also performed to assess cell proliferation/viability. Results Clear concentration-dependent increases in comet formation and γH2AX-foci/cell were noted for the TP53 MUT cells with no or lower increases being noted in the corresponding TP53 WT cells. Also, greater anti-proliferative and cell killing effects were noted in the TP53 MUT cells than in the TP53 WT cells. Conclusions This study's data suggests that P53 status/functioning is a key factor in determining the sensitivity of NSCLC and CRC cancer cells towards CHK1 inhibition, even in circumstances conducive to high replicative stress.
Collapse
Affiliation(s)
- Ali JN Duabil
- Leicester Cancer Research Centre, Department of Genetics & Genome Biology, University of Leicester, LE1 7RH Leics, UK
- Department of Surgery, Faculty of Medicine, University of Kufa, Najaf, Iraq
| | - Christian R Cooper
- Leicester Cancer Research Centre, Department of Genetics & Genome Biology, University of Leicester, LE1 7RH Leics, UK
- MRC Oxford Institute for Radiation Oncology, University of Oxford, OX3 7DQ Oxon, UK
| | - Esraa Aldujaily
- Leicester Cancer Research Centre, Department of Genetics & Genome Biology, University of Leicester, LE1 7RH Leics, UK
- Department of Pathology & Forensic Medicine, Faculty of Medicine, University of Kufa, Najaf, Iraq
| | - Sarah ER Halford
- Cancer Research UK Centre for Drug Development, London E20 1JQ, UK
| | | | | | - George DD Jones
- Leicester Cancer Research Centre, Department of Genetics & Genome Biology, University of Leicester, LE1 7RH Leics, UK
| |
Collapse
|
15
|
Zhao Y, Short NJ, Kantarjian HM, Chang TC, Ghate PS, Qu C, Macaron W, Jain N, Thakral B, Phillips AH, Khoury J, Garcia-Manero G, Zhang W, Fan Y, Yang H, Garris RS, Nasr LF, Kriwacki RW, Roberts KG, Konopleva M, Jabbour EJ, Mullighan CG. Genomic determinants of response and resistance to inotuzumab ozogamicin in B-cell ALL. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.06.23299616. [PMID: 38106221 PMCID: PMC10723521 DOI: 10.1101/2023.12.06.23299616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Inotuzumab ozogamicin (InO) is an antibody-drug conjugate that delivers calicheamicin to CD22-expressing cells. In a retrospective cohort of InO treated patients with B-cell acute lymphoblastic leukemia, we sought to understand the genomic determinants of response to InO. Acquired CD22 mutations were observed in 11% (3/27) of post-InO relapsed tumor samples. There were multiple CD22 mutations per sample and the mechanisms of CD22 escape included protein truncation, protein destabilization, and epitope alteration. Hypermutation by error-prone DNA damage repair (alternative end-joining, mismatch repair deficiency) drove CD22 escape. Acquired loss-of-function mutations in TP53 , ATM and CDKN2A were observed, suggesting compromise of the G1/S DNA damage checkpoint as a mechanism of evading InO-induced apoptosis. In conclusion, genetic alterations modulating CD22 expression and DNA damage response influence InO efficacy. The escape strategies within and beyond antigen loss to CD22-targeted therapy elucidated in this study provide insights into improving therapeutic approaches and overcoming resistance. KEY POINTS We identified multiple mechanisms of CD22 antigen escape from inotuzumab ozogamicin, including protein truncation, protein destabilization, and epitope alteration.Hypermutation caused by error-prone DNA damage repair was a driver of CD22 mutation and escape. VISUAL ABSTRACT
Collapse
|
16
|
Luo Y, Ni R, Jin X, Feng P, Dai C, Jiang L, Chen P, Yang L, Zhu Y. FOXD1 expression-based prognostic model for uveal melanoma. Heliyon 2023; 9:e21333. [PMID: 38027647 PMCID: PMC10651470 DOI: 10.1016/j.heliyon.2023.e21333] [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: 03/30/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
FOXD1, a new member of the FOX transcription factor family, serves as a mediator and biomarker for cell reprogramming. But its contribution to prognosis of uveal melanoma (UVM) is unclear. This study demonstrated that FOXD1 might promote tumor growth and invasion, because FOXD1 expression was negatively correlated with overall survival, progression-free survival, and disease-specific survival in UVM patients. This conjecture was verified in cell culture with human uveal melanoma cell line (MUM2B) as model cells. Additionally, the biological mechanisms of FOXD1 based on FOXD1-related genomic spectrum, molecular pathways, tumor microenvironment, and drug treatment sensitivity were examined using The Cancer Genome Atlas (TCGA) database, aiming to reasonably explain why FOXD1 leads to poor prognosis of UVM. On these bases, a novel tumor prognostic model was established using the FOXD1-related immunomodulators TMEM173, TNFRSF4, TNFSF13, and ULBP1, which will enable the stratification of disease seriousness and clinical treatment for patients.
Collapse
Affiliation(s)
- Yang Luo
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Renhao Ni
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Xiaojun Jin
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Peipei Feng
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Centre Lihuili Hospital, Ningbo, 315000, China
| | - Chenyi Dai
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Lingjing Jiang
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | | | - Lu Yang
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Yabin Zhu
- Health Science Center, Ningbo University, Ningbo, 315211, China
| |
Collapse
|
17
|
Alimbetov D, Umbayev B, Tsoy A, Begimbetova D, Davis T, Kipling D, Askarova S. Small molecule targeting of the p38/Mk2 stress signaling pathways to improve cancer treatment. BMC Cancer 2023; 23:895. [PMID: 37740222 PMCID: PMC10517462 DOI: 10.1186/s12885-023-11319-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: 03/14/2023] [Accepted: 08/18/2023] [Indexed: 09/24/2023] Open
Abstract
PURPOSE Although a long-term goal of cancer therapy always has been the development of agents that selectively destroy cancer cells, more recent trends have been to seek secondary agents that sensitize cancer cells to existing treatment regimens. In this regard, the present study explored the possibility of using small molecule inhibitors of p38MAPK/MK2 stress signaling pathways as potential agents to enhance the sensitivity of cancer cells with abrogated G1 checkpoint to the DNA damaging agent etoposide by specifically targeting the DNA damage-induced G2 cell cycle checkpoint. METHODS We have applied CCK8 and FACS-based viability assays and cell cycle analysis to investigate the effect of small molecules SB203580 and MK2.III on the sensitivity of small cell lung cancer cells (SCLC) that lack the G1 checkpoint to the DNA damaging agent Etoposide when used in combination. We have also assessed the effectiveness of combination chemotherapy on tumor xenograft suppression with etoposide and MK2.III in immunosuppressed mice. In addition, additional CCK8 cell viability analysis of the MDA-MB-231 breast cancer cell line, and SW620, and SW480 colorectal cancer cell lines was performed. RESULTS Results suggest that etoposide produces a profound effect on the cell cycle profile of cells in a manner that is consistent with the degree of cell viability that is seen using the viable cell assay. Results of the co-treatment experiments revealed that the p38/MK2 kinase inhibitors SB203580 and MK2.III both enhanced the DNA-damaging effects of etoposide on NCI-H69 cell viability in vitro. Results revealed that in vivo MK2.III was able to act as a chemosensitizer when used in combination with etoposide making NCI-H69 lung cancer cells sensitive to chemotherapeutic drug by 45% compared to single usage of the drug. We also report that MK2.III sensitizes metastatic cell lines SW-620 and MDA-MB-231 to etoposide but does not increase the sensitivity of non-metastasizing SW-480 colorectal cells to DNA damaging agent in vitro. CONCLUSION Findings reported in this study provide evidence that specific inhibitors of MK2 may indeed improve overall cancer therapy; however, their effectiveness depends on cell types.
Collapse
Affiliation(s)
- D Alimbetov
- Creehey Children's Cancer Research Institute, UT Health at San Antonio, San Antonio, USA.
| | - B Umbayev
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | - A Tsoy
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | - D Begimbetova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | - T Davis
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, UK
| | - D Kipling
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, UK
| | - Sh Askarova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan.
| |
Collapse
|
18
|
兰 玉, 王 凯, 蓝 智, 周 何, 孙 剑. [Dealcoholized red wine inhibits occurrence and progression of hepatocellular carcinoma possibly by inducing cell cycle arrest and apoptosis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:1297-1305. [PMID: 37712265 PMCID: PMC10505577 DOI: 10.12122/j.issn.1673-4254.2023.08.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Indexed: 09/16/2023]
Abstract
OBJECTIVE To investigate the inhibitory effect of dealcoholized red wine (DRW) on occurrence and progression of hepatocellular carcinoma (HCC) and explore its possible mechanisms. METHODS Three HCC cell lines (Huh7, HepG2 and SKHep-1) treated with 5, 10, 25, 50 and 100 μL/mL DRW were examined for changes in proliferation and colony formation ability using CCK-8 assay and colony formation assay. A nude mouse model bearing subcutaneous HCC xenograft was used to test the effect of 300 μL/day DRW for 4 weeks on tumor growth. The inhibitory effect of 300 μL/day DRW for 6 weeks on tumor growth was also observed in a mouse model of chemically induced HCC by examining the tumor number, largest tumor diameter and the liver/body ratio. RNA-seq technique was used for transcriptome sequencing of Huh7 cells treated with DRW (75 μL/mL) for 48 h, and gene-set enrichment analysis (GSEA) was performed to identify the changes in genes and pathways. Flow cytometry assay was used to analyze the changes in cell cycle and apoptosis of the cells. RESULTS DRW inhibited the proliferation of the HCC cell lines in a concentration-and time-dependent manner, and concentration-dependently inhibited colony formation of the cells. Treatment with DRW significantly reduced the volume of subcutaneous tumor xenograft in the tumor-bearing nude mice (P < 0.05), and lowered the number of tumors (P < 0.001), the largest tumor diameter (P < 0.05) and the liver/body ratio (P < 0.01) in mice with chemically induced HCC. RNA-seq showed that 634 genes were significantly up-regulated and 478 were down-regulated in Huh7 cells after treatment with DRW. Gene-set enrichment analysis revealed that DRW significantly down-regulated cell cycle-related pathways (E2F Targets, G2M Checkpoint and MYC Targets) and up-regulated apoptosis pathways. Flow cytometry assay showed that DRW induced cell cycle arrest in G1 phase and apoptosis of Huh7 cells. CONCLUSION DRW inhibits the occurrence and progression of HCC, and this effect is mediated possibly by inducing cell cycle arrest and apoptosis.
Collapse
Affiliation(s)
- 玉 兰
- />器官衰竭防治国家重点实验室,广东省病毒性肝炎研究重点实验室,南方医科大学南方医院感染内科,广东 广州 510515State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 凯风 王
- />器官衰竭防治国家重点实验室,广东省病毒性肝炎研究重点实验室,南方医科大学南方医院感染内科,广东 广州 510515State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 智贤 蓝
- />器官衰竭防治国家重点实验室,广东省病毒性肝炎研究重点实验室,南方医科大学南方医院感染内科,广东 广州 510515State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 何琪 周
- />器官衰竭防治国家重点实验室,广东省病毒性肝炎研究重点实验室,南方医科大学南方医院感染内科,广东 广州 510515State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 剑 孙
- />器官衰竭防治国家重点实验室,广东省病毒性肝炎研究重点实验室,南方医科大学南方医院感染内科,广东 广州 510515State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
19
|
Gan C, Li W, Xu J, Pang L, Tang L, Yu S, Li A, Ge H, Huang R, Cheng H. Advances in the study of the molecular biological mechanisms of radiation-induced brain injury. Am J Cancer Res 2023; 13:3275-3299. [PMID: 37693137 PMCID: PMC10492106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 07/12/2023] [Indexed: 09/12/2023] Open
Abstract
Radiation therapy is one of the most commonly used treatments for head and neck cancers, but it often leads to radiation-induced brain injury. Patients with radiation-induced brain injury have a poorer quality of life, and no effective treatments are available. The pathogenesis of this condition is unknown. This review summarizes the molecular biological mechanism of radiation-induced brain injury and provides research directions for future studies. The molecular mechanisms of radiation-induced brain injury are diverse and complex. Radiation-induced chronic neuroinflammation, destruction of the blood-brain barrier, oxidative stress, neuronal damage, and physiopathological responses caused by specific exosome secretion lead to radiation-induced brain injury.
Collapse
Affiliation(s)
- Chen Gan
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Wen Li
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Jian Xu
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Lulian Pang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Lingxue Tang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Sheng Yu
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Anlong Li
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Han Ge
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Runze Huang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Huaidong Cheng
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Shenzhen Hospital of Southern Medical UniversityShenzhen, Guangdong, China
| |
Collapse
|
20
|
Moloudi K, Abrahamse H, George BP. Photodynamic therapy induced cell cycle arrest and cancer cell synchronization: review. Front Oncol 2023; 13:1225694. [PMID: 37503319 PMCID: PMC10369002 DOI: 10.3389/fonc.2023.1225694] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 06/21/2023] [Indexed: 07/29/2023] Open
Abstract
Cell cycle arrest (CCA) is seen as a prime candidate for effective cancer therapy. This mechanism can help researchers to create new treatments to target cancer cells at particular stages of the cell cycle (CC). The CCA is a characteristic of various therapeutic modalities, including radiation (RT) and chemotherapy (CT), which synchronizes the cells and facilitates the standardization of radio-chemotherapy protocols. Although it was discovered that photodynamic treatment (PDT) had a biological effect on CCA in cancer cells, the mechanism remains unclear. Furthermore, besides conventional forms of cell death such as apoptosis, autophagy, and necrosis, various unconventional types of cell death including pyroptosis, mitotic catastrophe, paraptosis, ferroptosis, necroptosis, and parthanatos after PDT have been reported. Thus, a variety of elements, such as oxygen, the tumor's microenvironment, the characteristics of light, and photosensitizer (PS), influence the effectiveness of the PDT treatment, which have not yet been studied clearly. This review focuses on CCA induced by PDT for a variety of PSs agents on various cell lines. The CCA by PDT can be viewed as a remarkable effect and instructive for the management of the PDT protocol. Regarding the relationship between the quantity of reactive oxygen species (ROS) and its biological consequences, we have proposed two mathematical models in PDT. Finally, we have gathered recent in vitro and in vivo studies about CCA post-PDT at various stages and made suggestions about how it can standardize, potentiate, and customize the PDT methodology.
Collapse
|
21
|
Tian L, Li C, Xiang L, Zeng J, Chen S, Guo W, Chen S, Wang Y, He X, Su P, Xu C. T52 attenuates oncogenic STAT3 signaling and suppresses osteosarcoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154799. [PMID: 37058945 DOI: 10.1016/j.phymed.2023.154799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/12/2023] [Accepted: 04/01/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND T52 is a steroidal saponin extracted from the traditional Chinese herb Rohdea fargesii (Baill.), and it is reported to possess strong anti-proliferative capabilities in human pharyngeal carcinoma cell lines. However, whether T52 has anti-osteosarcoma properties, and its potential mechanism is remains unknown. PURPOSE To examine the outcome and underlying mechanism of T52 in osteosarcomas (OS). METHODS/STUDY DESIGNS The physiological roles of T52 in OS cells were examined using CCK-8, colony formation (CF), EdU staining, cell cycle/apoptosis and cell migration/invasion assays. The relevant T52 targets against OS were assessed via bioinformatics prediction, and the binding sites were analyzed by molecular docking. Western blot analysis was carried out to examine the levels of factors associated with apoptosis, cell cycle, and STAT3 signaling pathway activation. RESULTS T52 markedly diminished the proliferation, migration, and invasion of OS cells, and promoted G2/M arrest and apoptosis in a dose-dependent fashion (DDF) in vitro. Mechanistically, molecular docking predicted that T52 stably associated with STAT3 Src homology 2 (SH2) domain residues. Western blot revealed that T52 suppressed the STAT3 signaling pathway, as well as the expression of the downstream targets, such as, Bcl-2, Cyclin D1, and c-Myc. In addition, the anti-OS property of T52 were partially reversed by STAT3 reactivation, which confirmed that STAT3 signaling is critical for regulating the anti-OS property of T52. CONCLUSION We firstly demonstrated that T52 possessed strong anti-osteosarcoma property in vitro, which was brought on by the inhibition of the STAT3 signaling pathway. Our findings provided pharmacological support for treating OS with T52.
Collapse
Affiliation(s)
- Liru Tian
- Research Center for Translational Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510006, China
| | - Chuan Li
- Research Center for Translational Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510006, China
| | - Limin Xiang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou 510006, China
| | - Jia Zeng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou 510006, China
| | - Shuqing Chen
- Department of Traditional Chinese Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510006, China
| | - Weimin Guo
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Shulin Chen
- Research Center for Translational Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510006, China; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yihai Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou 510006, China
| | - Xiangjiu He
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou 510006, China.
| | - Peiqiang Su
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; The Department of Orthopedics, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road 2, Guangzhou, 510080, China.
| | - Caixia Xu
- Research Center for Translational Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
22
|
Lin ES, Huang YH, Chung JC, Su HH, Huang CY. The Inhibitory Effects and Cytotoxic Activities of the Stem Extract of Nepenthes miranda against Single-Stranded DNA-Binding Protein and Oral Carcinoma Cells. PLANTS (BASEL, SWITZERLAND) 2023; 12:plants12112188. [PMID: 37299167 DOI: 10.3390/plants12112188] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023]
Abstract
The carnivorous pitcher plants of the genus Nepenthes exhibit many ethnobotanical uses, including treatments of stomachache and fever. In this study, we prepared different extracts from the pitcher, stem, and leaf extracts of Nepenthes miranda obtained using 100% methanol and analyzed their inhibitory effects on recombinant single-stranded DNA-binding protein (SSB) from Klebsiella pneumoniae (KpSSB). SSB is essential for DNA replication and cell survival and thus an attractive target for potential antipathogen chemotherapy. Different extracts prepared from Sinningia bullata, a tuberous member of the flowering plant family Gesneriaceae, were also used to investigate anti-KpSSB properties. Among these extracts, the stem extract of N. miranda exhibited the highest anti-KpSSB activity with an IC50 value of 15.0 ± 1.8 μg/mL. The cytotoxic effects of the stem extract of N. miranda on the survival and apoptosis of the cancer cell lines Ca9-22 gingival carcinoma, CAL27 oral adenosquamous carcinoma, PC-9 pulmonary adenocarcinoma, B16F10 melanoma, and 4T1 mammary carcinoma cells were also demonstrated and compared. Based on collective data, the cytotoxic activities of the stem extract at a concentration of 20 μg/mL followed the order Ca9-22 > CAL27 > PC9 > 4T1 > B16F10 cells. The stem extract of N. miranda at a concentration of 40 μg/mL completely inhibited Ca9-22 cell migration and proliferation. In addition, incubation with this extract at a concentration of 20 μg/mL boosted the distribution of the G2 phase from 7.9% to 29.2% in the Ca9-22 cells; in other words, the stem extract might suppress Ca9-22 cell proliferation by inducing G2 cell cycle arrest. Through gas chromatography-mass spectrometry, the 16 most abundant compounds in the stem extract of N. miranda were tentatively identified. The 10 most abundant compounds in the stem extract of N. miranda were used for docking analysis, and their docking scores were compared. The binding capacity of these compounds was in the order sitosterol > hexadecanoic acid > oleic acid > plumbagin > 2-ethyl-3-methylnaphtho[2,3-b]thiophene-4,9-dione > methyl α-d-galactopyranoside > 3-methoxycatechol > catechol > pyrogallol > hydroxyhydroquinone; thus, sitosterol might exhibit the greatest inhibitory capacity against KpSSB among the selected compounds. Overall, these results may indicate the pharmacological potential of N. miranda for further therapeutic applications.
Collapse
Affiliation(s)
- En-Shyh Lin
- Department of Beauty Science, National Taichung University of Science and Technology, Taichung City 403, Taiwan
| | - Yen-Hua Huang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan
| | - Jo-Chi Chung
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan
| | - Hsin-Hui Su
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan City 717, Taiwan
| | - Cheng-Yang Huang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung City 402, Taiwan
| |
Collapse
|
23
|
Kang Y, Zhang Q, Feng YX, Yang L, Yu XZ. Exogenous proline activated an integrated response of NER and HR pathways to reduce DNA damage in rice seedlings under chromium stress. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:51792-51803. [PMID: 36820975 DOI: 10.1007/s11356-023-26009-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/14/2023] [Indexed: 06/18/2023]
Abstract
The DNA damage induced by hexavalent chromium [Cr(VI)] pollutant causes a genotoxic effect on rice seedlings. Hereby, we examined the effects of exogenous proline (Pro) on the alleviation of DNA damage in rice seedlings under different effective concentrations of Cr(VI). Our results revealed that Cr(VI) stress induced reactive oxygen species (ROS), i.e., H2O2 and O2·- accumulation in rice seedlings, repressed genes expression activated in the homologous recombination (HR) and nucleotide excision repair (NER) pathways, and caused DNA damage. Exogenous application of Pro increased Cr accumulation in rice roots, but decreased Cr accumulation in rice shoots, wherein Pro application decreased ROS accumulation in both tissues of rice seedlings. The comet assays suggested that exogenous application of Pro significantly alleviated the DNA damage in rice seedlings during Cr(VI) treatments, judged by the Olive tail moment and tail DNA. Transcriptional assays revealed that exogenous Pro upregulated the expression level of genes associated with the HR and NER pathways and triggered coordinated actions of both repairing pathways to modulate DNA lesion in rice plants during exposure to Cr(VI). Calculations from gene expression variation factors showed that regulative effect of exogenous application of Pro on DNA repair pathways was highly activated at 2.0 mg Cr/L. The current study revealed that Cr(VI) affect rice plants and exogenous Pro rescue these effects by the activation of HR and NER pathways.
Collapse
Affiliation(s)
- Yi Kang
- The Guangxi Key Laboratory of Theory & Technology for Environmental Pollution Control, College of Environmental Science & Engineering, Guilin University of Technology|, Guilin, 541004, People's Republic of China
| | - Qing Zhang
- The Guangxi Key Laboratory of Theory & Technology for Environmental Pollution Control, College of Environmental Science & Engineering, Guilin University of Technology|, Guilin, 541004, People's Republic of China
| | - Yu-Xi Feng
- The Guangxi Key Laboratory of Theory & Technology for Environmental Pollution Control, College of Environmental Science & Engineering, Guilin University of Technology|, Guilin, 541004, People's Republic of China
| | - Li Yang
- The Guangxi Key Laboratory of Theory & Technology for Environmental Pollution Control, College of Environmental Science & Engineering, Guilin University of Technology|, Guilin, 541004, People's Republic of China
| | - Xiao-Zhang Yu
- The Guangxi Key Laboratory of Theory & Technology for Environmental Pollution Control, College of Environmental Science & Engineering, Guilin University of Technology|, Guilin, 541004, People's Republic of China.
| |
Collapse
|
24
|
Schutte T, Embaby A, Steeghs N, van der Mierden S, van Driel W, Rijlaarsdam M, Huitema A, Opdam F. Clinical development of WEE1 inhibitors in gynecological cancers: A systematic review. Cancer Treat Rev 2023; 115:102531. [PMID: 36893690 DOI: 10.1016/j.ctrv.2023.102531] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/24/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023]
Abstract
INTRODUCTION The anti-tumor activity of WEE1 inhibitors (WEE1i) in gynecological malignancies has recently been demonstrated in clinical trials and its rationale is based on biological/molecular features of gynecological cancers. With this systematic review, we aim to outline the clinical development and current evidence regarding the efficacy and safety of these targeted agents in in this patient group. METHODS Systematic literature review of trials including patients with gynecological cancers treated with a WEE1i. The primary objective was to summarize the efficacy of WEE1i in gynecological malignancies regarding objective response rate (ORR), clinical benefit rate (CBR), overall survival (OS) and progression-free survival (PFS). Secondary objectives included toxicity profile, Maximum Tolerated Dose (MTD), pharmacokinetics, drug-drug interactions and exploratory objectives such as biomarkers for response. RESULTS 26 records were included for data extraction. Almost all trials used the first-in-class WEE1i adavosertib; one conference abstract reported about Zn-c3. The majority of the trials included diverse solid tumors (n = 16). Six records reported efficacy results of WEE1i in gynecological malignancies (n = 6). Objective response rates of adavosertib monotherapy or in combination with chemotherapy ranged between 23% and 43% in these trials. Median PFS ranged from 3.0 to 9.9 months. The most common adverse events were bone marrow suppression, gastrointestinal toxicities and fatigue. Mainly alterations in cell cycle regulator genes TP53 and CCNE1 were potential predictors of response. CONCLUSION This report summarizes encouraging clinical development of WEE1i in gynecological cancers and considers its application in future studies. Biomarker-driven patient selection might be essential to increase the response rates.
Collapse
Affiliation(s)
- Tim Schutte
- Department of Internal Medicine and Department of Medical Oncology, Amsterdam UMC, Location VUmc, Amsterdam, Netherlands.
| | - Alaa Embaby
- Department of Clinical Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, Netherlands; Department of Medical Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, Netherlands
| | - Stevie van der Mierden
- Scientific Information Service, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Willemien van Driel
- Department of Gynecological Oncology, The Netherlands Cancer Insitute - Antoni van Leeuwenhoek, Amsterdam, Netherlands
| | - Martin Rijlaarsdam
- Department of Medical Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, Netherlands
| | - Alwin Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, Netherlands; Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands; Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Frans Opdam
- Department of Medical Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, Netherlands
| |
Collapse
|
25
|
Chen PJ, Lin ES, Su HH, Huang CY. Cytotoxic, Antibacterial, and Antioxidant Activities of the Leaf Extract of Sinningia bullata. PLANTS (BASEL, SWITZERLAND) 2023; 12:859. [PMID: 36840206 PMCID: PMC9967939 DOI: 10.3390/plants12040859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 06/12/2023]
Abstract
Sinningia bullata is a tuberous member of the flowering plant family Gesneriaceae. Prior to this work, the antibacterial, antioxidant, and cytotoxic properties of S. bullata were undetermined. Here, we prepared different extracts from the leaf, stem, and tuber of S. bullata and investigated their pharmacological activities. The leaf extract of S. bullata, obtained by 100% acetone (Sb-L-A), had the highest total flavonoid content, antioxidation capacity, and cytotoxic and antibacterial activities. Sb-L-A displayed a broad range of antibacterial activities against Escherichia coli, Staphylococcus aureus, and Pseudomonas aeruginosa. The inhibition zones of Sb-L-A ranged from 8 to 30 mm and were in the following order: S. aureus > E. coli > P. aeruginosa. Incubation of B16F10 melanoma cells with Sb-L-A at a concentration of 80 μg/mL caused deaths at the rate of 96%, reduced migration by 100%, suppressed proliferation and colony formation by 99%, and induced apoptosis, which was observed in 96% of the B16F10 cells. In addition, the cytotoxic activities of Sb-L-A were synergistically enhanced when coacting with the antitumor drug epothilone B. Sb-L-A was also used to determine the cytotoxic effects against 4T1 mammary carcinoma cells. Sb-L-A of 60 μg/mL boosted the distribution of the G2 phase from 1.4% to 24.4% in the B16F10 cells. Accordingly, Sb-L-A might suppress melanoma cell proliferation by inducing G2 cell-cycle arrest. The most abundant compounds in Sb-L-A were identified using gas chromatography-mass spectrometry. Overall, the collective data in this study may indicate the pharmacological potentials of Sb-L-A for possible medical applications.
Collapse
Affiliation(s)
- Pin-Jui Chen
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan;
| | - En-Shyh Lin
- Department of Beauty Science, National Taichung University of Science and Technology, Taichung City 403, Taiwan;
| | - Hsin-Hui Su
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan City 717, Taiwan;
| | - Cheng-Yang Huang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung City 402, Taiwan
| |
Collapse
|
26
|
Zhang H, Yao G, He M. Transcriptome analysis of gene expression profiling from the deep sea in situ to the laboratory for the cold seep mussel Gigantidas haimaensis. BMC Genomics 2022; 23:828. [DOI: 10.1186/s12864-022-09064-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022] Open
Abstract
Abstract
Background
The deep-sea mussel Gigantidas haimaensis is a representative species from the Haima cold seep ecosystem in the South China Sea that establishes endosymbiosis with chemotrophic bacteria. During long-term evolution, G. haimaensis has adapted well to the local environment of cold seeps. Until now, adaptive mechanisms responding to environmental stresses have remained poorly understood.
Results
In this study, transcriptomic analysis was performed for muscle tissue of G. haimaensis in the in situ environment (MH) and laboratory environment for 0 h (M0), 3 h (M3) and 9 h (M9), and 187,368 transcript sequences and 22,924 annotated differentially expressed genes (DEGs) were generated. Based on Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, these DEGs were enriched with a broad spectrum of biological processes and pathways, including those associated with antioxidants, apoptosis, chaperones, immunity and metabolism. Among these significantly enriched pathways, protein processing in the endoplasmic reticulum and metabolism were the most affected metabolic pathways. These results may imply that G. haimaensis struggles to support the life response to environmental change by changing gene expression profiles.
Conclusion
The present study provides a better understanding of the biological responses and survival strategies of the mussel G. haimaensis from deep sea in situ to the laboratory environment.
Collapse
|
27
|
Wang M, Li L, Fan T, Cao L, Zhang J, Li S, Liu C, Liu X. Semisynthetic aurones A14 protects against T-cell acute lymphoblastic leukemia via suppressing proliferation and inducing cell cycle arrest with apoptosis. Chin Med 2022; 17:137. [PMID: 36510253 PMCID: PMC9743678 DOI: 10.1186/s13020-022-00693-6] [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: 10/04/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Acute lymphoblastic leukemia is an aggressive neoplasm and seriously threatens human health. A14 is one kind of semisynthetic aurone that exhibits the capability to inhibit prostate cancer, but little is known about the role of A14 on T-cell acute lymphoblastic leukemia. METHODS Firstly, the effects of A14 on the ability of leukemia cells to proliferate were measured by Vi-cell counter. Then, we detected the cell cycle and apoptosis by flow cytometry and characterized the related protein expression using immunoblotting. In addition, we constructed stable luciferase expressing cell lines for use in a cell derived xenograft mouse model to measure the effect of A14 on T-cell acute lymphoblastic leukemia. RESULTS Results exhibited that A14 markedly suppressed cell proliferation and induced G2/M phase arrest along with cell cycles regulating proteins changes. A14 led to apoptosis in leukemia cells, at least partly, through the cytochrome c signaling pathway. Experiments in cell derived xenograft mouse model also showed that A14 markedly ameliorated the survival rate. CONCLUSIONS The present study revealed that semisynthetic aurones A14 can effectively protect against T-cell acute lymphoblastic leukemia progression both in vitro and in vivo, indicating the capability of A14 as a promising drug for the treatment of T-cell acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- Meng Wang
- grid.256884.50000 0004 0605 1239Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Lisi Li
- grid.256884.50000 0004 0605 1239Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Tengyun Fan
- grid.256884.50000 0004 0605 1239Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Lixue Cao
- grid.256884.50000 0004 0605 1239Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Jiayi Zhang
- grid.256884.50000 0004 0605 1239Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Shuang Li
- grid.256884.50000 0004 0605 1239Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Chunming Liu
- grid.266539.d0000 0004 1936 8438Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Xifu Liu
- grid.256884.50000 0004 0605 1239Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, College of Life Science, Hebei Normal University, Shijiazhuang, China ,Jianyuan Science & Technology (Zhangjiakou) Co., Ltd., Zhangjiakou, China
| |
Collapse
|
28
|
Drug Repurposing Applications to Overcome Male Predominance via Targeting G2/M Checkpoint in Human Esophageal Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14235854. [PMID: 36497337 PMCID: PMC9741366 DOI: 10.3390/cancers14235854] [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: 10/10/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/29/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is strongly characterized by a male predominance with higher mortality rates and worse responses to treatment in males versus females. Despite the role of sex hormones, other causes that may contribute to sex bias in ESCC remain largely unknown, especially as age increases and the hormone difference begins to diminish between sexes. In this study, we analyzed genomics, transcriptomics, and epigenomics from 663 ESCC patients and found that G2/M checkpoint pathway-related sex bias and age bias were significantly present in multi-omics data. In accordance with gene expression patterns across sexes, ten compounds were identified by applying drug repurposing from three drug sensitivity databases: The Connective Map (CMap), Genomics of Drug Sensitivity in Cancer (GDSC), and The Cancer Therapeutic Response Portal (CTRP). MK1775 and decitabine showed better efficacy in two male ESCC cell lines in vitro and in vivo. The drugs' relevance to the transition between G2 and M was especially evident in male cell lines. In our study, we first validated the sex bias of the G2/M checkpoint pathway in ESCC and then determined that G2/M targets may be included in combination therapy for male patients to improve the efficacy of ESCC treatment.
Collapse
|
29
|
Liu HW, Chiang WY, Huang YH, Huang CY. The Inhibitory Effects and Cytotoxic Activities of the Stem Extract of Sarracenia purpurea against Melanoma Cells and the SsbA Protein. PLANTS (BASEL, SWITZERLAND) 2022; 11:plants11223164. [PMID: 36432892 PMCID: PMC9692666 DOI: 10.3390/plants11223164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/17/2022] [Accepted: 11/17/2022] [Indexed: 06/12/2023]
Abstract
The Staphylococcus aureus SsbA protein (SaSsbA) is a single-stranded DNA-binding protein (SSB) that is categorically required for DNA replication and cell survival, and it is thus an attractive target for potential antipathogen chemotherapy. In this study, we prepared the stem extract of Sarracenia purpurea obtained from 100% acetone to investigate its inhibitory effect against SaSsbA. In addition, the cytotoxic effects of this extract on the survival, apoptosis, proliferation, and migration of B16F10 melanoma cells were also examined. Initially, myricetin, quercetin, kaempferol, dihydroquercetin, dihydrokaempferol, rutin, catechin, β-amyrin, oridonin, thioflavin T, primuline, and thioflavin S were used as possible inhibitors against SaSsbA. Of these compounds, dihydrokaempferol and oridonin were capable of inhibiting the ssDNA-binding activity of SaSsbA with respective IC50 values of 750 ± 62 and 2607 ± 242 μM. Given the poor inhibition abilities of dihydrokaempferol and oridonin, we screened the extracts of S. purpurea, Nepenthes miranda, and Plinia cauliflora for SaSsbA inhibitors. The stem extract of S. purpurea exhibited high anti-SaSsbA activity, with an IC50 value of 4.0 ± 0.3 μg/mL. The most abundant compounds in the stem extract of S. purpurea were identified using gas chromatography−mass spectrometry. The top five most abundant contents in this extract were driman-8,11-diol, deoxysericealactone, stigmast-5-en-3-ol, apocynin, and α-amyrin. Using the MOE-Dock tool, the binding modes of these compounds, as well as dihydrokaempferol and oridonin, to SaSsbA were elucidated, and their binding energies were also calculated. Based on the S scores, the binding capacity of these compounds was in the following order: deoxysericealactone > dihydrokaempferol > apocynin > driman-8,11-diol > stigmast-5-en-3-ol > oridonin > α-amyrin. Incubation of B16F10 cells with the stem extract of S. purpurea at a concentration of 100 μg/mL caused deaths at the rate of 76%, reduced migration by 95%, suppressed proliferation and colony formation by 99%, and induced apoptosis, which was observed in 96% of the B16F10 cells. Overall, the collective data in this study indicate the pharmacological potential of the stem extract of S. purpurea for further medical applications.
Collapse
Affiliation(s)
- Hong-Wen Liu
- Department of Rheumatology and Immunology, Antai Medical Care Corporation Antai Tian-Sheng Memorial Hospital, Pingtung 928, Taiwan
| | - Wei-Yu Chiang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan
| | - Yen-Hua Huang
- Department of Rheumatology and Immunology, Antai Medical Care Corporation Antai Tian-Sheng Memorial Hospital, Pingtung 928, Taiwan
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan
| | - Cheng-Yang Huang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung City 402, Taiwan
| |
Collapse
|
30
|
Zhu LY, Yuan JB, Zhang L, He CX, Lin X, Xu B, Jin GH. Loss of MLL Induces Epigenetic Dysregulation of Rasgrf1 to Attenuate Kras-Driven Lung Tumorigenesis. Cancer Res 2022; 82:4153-4163. [PMID: 36098964 DOI: 10.1158/0008-5472.can-22-1475] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/01/2022] [Accepted: 09/09/2022] [Indexed: 12/14/2022]
Abstract
Menin is necessary for the formation of the menin/mixed lineage leukemia (MLL) complex and is recruited directly to chromatin. Menin is an important tumor suppressor in several cancer types, including lung cancer. Here, we investigated the role of MLL in menin-regulated lung tumorigenesis. Ablation of MLL suppressed KrasG12D-induced lung tumorigenesis in a genetically engineered mouse model. MLL deficiency decreased histone H3 lysine 4 trimethylation (H3K4me3) and subsequently suppressed expression of the Ras protein-specific guanine nucleotide-releasing factor 1 (Rasgrf1) gene. Rasgrf1 was essential for the GTP-bound active state of Kras and the activation of Kras downstream pathways as well as their cancer-promoting activities. MI-3, a small-molecule inhibitor targeting MLL, specifically inhibited the growth of Kras-mutated lung cancer cells in vitro and in vivo with minimal effect on wild-type Kras lung cancer growth. Together, these results demonstrate a novel tumor promoter function of MLL in mutant Kras-induced lung tumorigenesis and further indicate that specific blockade of the MLL-Rasgrf1 pathway may be a potential therapeutic strategy for the treatment of tumors containing Kras mutations. SIGNIFICANCE Activation of mutant Kras is dependent on MLL-mediated epigenetic regulation of Rasgrf1, conferring sensitivity to small-molecule inhibition of MLL in Kras-driven lung cancer.
Collapse
Affiliation(s)
- Ling-Yu Zhu
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, Fujian, P.R. China
| | - Jun-Bo Yuan
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, Fujian, P.R. China
| | - Li Zhang
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, Fujian, P.R. China
| | - Chun-Xiao He
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, Fujian, P.R. China
| | - Xiao Lin
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, Fujian, P.R. China
| | - Bin Xu
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, Fujian, P.R. China
| | - Guang-Hui Jin
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, Fujian, P.R. China.,State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, P.R. China
| |
Collapse
|
31
|
Chen C, Wang Y, Hu MQ, Li H, Chen X, Qiang G, Sun Y, Zhu Y, Li B. Discovery of pyrrolo[2,3-d]pyrimidine-based molecules as a Wee1 inhibitor template. Bioorg Med Chem Lett 2022; 75:128973. [PMID: 36075370 DOI: 10.1016/j.bmcl.2022.128973] [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: 05/04/2022] [Revised: 08/12/2022] [Accepted: 08/29/2022] [Indexed: 11/02/2022]
Abstract
In the past decade, Wee1 inhibition has received widespread attention as a cancer therapy. Our research aims to discover effective, selective and drug-like Wee1 inhibitors. Herein, a series of compounds with pyrrolo[2,3-d]pyrimidine-based heterocycles were designed, synthesized and confirmed to inhibit Wee1 kinase. The inhibitors afforded good potency in Wee1 Kinase inhibitory activity in enzymatic assays. These compounds showed strong proliferation inhibition against NCI-1299 cell lines and had acceptable pharmacokinetic properties. These derivatives are promising inhibitors that warrant further evaluation, towards the development of potential anticancer drug.
Collapse
Affiliation(s)
- Changjun Chen
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China; Medicinal Chemistry Department, Shouyao Holdings (Beijing) Co., Ltd., Beijing, China
| | - Yeliu Wang
- Medicinal Chemistry Department, Shouyao Holdings (Beijing) Co., Ltd., Beijing, China
| | - Min-Qi Hu
- Medicinal Chemistry Department, Shouyao Holdings (Beijing) Co., Ltd., Beijing, China
| | - Hongjuan Li
- Discovery Biology Department, Shouyao Holdings (Beijing) Co., Ltd., Beijing, China
| | - Xi Chen
- Medicinal Chemistry Department, Shouyao Holdings (Beijing) Co., Ltd., Beijing, China
| | - Gan Qiang
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing,China
| | - Yinghui Sun
- Discovery Biology Department, Shouyao Holdings (Beijing) Co., Ltd., Beijing, China
| | - Yan Zhu
- Medicinal Chemistry Department, Shouyao Holdings (Beijing) Co., Ltd., Beijing, China.
| | - Binghui Li
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
| |
Collapse
|
32
|
Przystupski D, Ussowicz M. Landscape of Cellular Bioeffects Triggered by Ultrasound-Induced Sonoporation. Int J Mol Sci 2022; 23:ijms231911222. [PMID: 36232532 PMCID: PMC9569453 DOI: 10.3390/ijms231911222] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
Sonoporation is the process of transient pore formation in the cell membrane triggered by ultrasound (US). Numerous studies have provided us with firm evidence that sonoporation may assist cancer treatment through effective drug and gene delivery. However, there is a massive gap in the body of literature on the issue of understanding the complexity of biophysical and biochemical sonoporation-induced cellular effects. This study provides a detailed explanation of the US-triggered bioeffects, in particular, cell compartments and the internal environment of the cell, as well as the further consequences on cell reproduction and growth. Moreover, a detailed biophysical insight into US-provoked pore formation is presented. This study is expected to review the knowledge of cellular effects initiated by US-induced sonoporation and summarize the attempts at clinical implementation.
Collapse
|
33
|
High DHCR7 Expression Predicts Poor Prognosis for Cervical Cancer. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:8383885. [PMID: 36164611 PMCID: PMC9508458 DOI: 10.1155/2022/8383885] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 07/19/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022]
Abstract
DHCR7 is a rate-limiting enzyme in cholesterol synthesis. The expression pattern and prognostic value of DHCR7 in cervical cancer are unknown. We investigated the relationship between DHCR7 expression and clinicopathological features of cervical cancer patients. The dataset was acquired from TCGA database. The Wilcoxon rank sum test was used to explore DHCR7 expression level in cervical cancer. The Kruskal-Wallis test and the logistic regression were performed to estimate the association between the DHCR7 and clinical features. The Kaplan-Meier and Cox regression analyses were used to evaluate factors that affect cervical cancer prognosis. GSEA was used to screen the DHCR7-related pathways. We found that DHCR7 was increased in cervical cancer samples and increased DHCR7 was correlated with advanced T stage, lymph node invasion, and clinical stage (P < 0.05). Patients with elevated DHCR7 levels had poorer overall survival (P = 0.021), progression-free interval (P = 0.002), and disease-specific survival (P = 0.005). Cox analysis revealed that DHCR7 was an independent prognostic factor in cervical cancer (P = 0.005). WNT activated receptor activity, G2/M checkpoint, mTORC1 signaling, KRAS signaling, regulation of cholesterol biosynthetic, FGF signaling, T-cell receptor signaling, JAK/STAT signaling cascade T cell activation, and macrophage migration were enriched in high DHCR7 phenotype. Our data also showed that DHCR7 moderately correlates with T-cell infiltration, including CD8+ T-cells. Conclusion. Increased DHCR7 expression is associated with poor survival in cervical cancer.
Collapse
|
34
|
Wang M, Chen X, Tan P, Wang Y, Pan X, Lin T, Jiang Y, Wang B, Xu H, Wang Y, Yang Y, Wang J, Zhao L, Zhang J, Zhong A, Peng Y, Du J, Zhang Q, Zheng J, Chen J, Dai S, Na F, Lu Z, Liu J, Zheng X, Yang L, Zhang P, Han P, Gong Q, Zhong Q, Xiao K, Yang H, Deng H, Zhao Y, Shi H, Man J, Gou M, Zhao C, Dai L, Xue Z, Chen L, Wang Y, Zeng M, Huang C, Wei Q, Wei Y, Liu Y, Chen C. Acquired semi-squamatization during chemotherapy suggests differentiation as a therapeutic strategy for bladder cancer. Cancer Cell 2022; 40:1044-1059.e8. [PMID: 36099882 DOI: 10.1016/j.ccell.2022.08.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 05/16/2022] [Accepted: 08/11/2022] [Indexed: 02/06/2023]
Abstract
Cisplatin-based chemotherapy remains the primary treatment for unresectable and metastatic muscle-invasive bladder cancers (MIBCs). However, tumors frequently develop chemoresistance. Here, we established a primary and orthotopic MIBC mouse model with gene-edited organoids to recapitulate the full course of chemotherapy in patients. We found that partial squamous differentiation, called semi-squamatization, is associated with acquired chemoresistance in both mice and human MIBCs. Multi-omics analyses showed that cathepsin H (CTSH) is correlated with chemoresistance and semi-squamatization. Cathepsin inhibition by E64 treatment induces full squamous differentiation and pyroptosis, and thus specifically restrains chemoresistant MIBCs. Mechanistically, E64 treatment activates the tumor necrosis factor pathway, which is required for the terminal differentiation and pyroptosis of chemoresistant MIBC cells. Our study revealed that semi-squamatization is a type of lineage plasticity associated with chemoresistance, suggesting that differentiation via targeting of CTSH is a potential therapeutic strategy for the treatment of chemoresistant MIBCs.
Collapse
Affiliation(s)
- Manli Wang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xuelan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ping Tan
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yiyun Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiangyu Pan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tianhai Lin
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yong Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Bo Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Huan Xu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuying Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yucen Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jian Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lei Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiapeng Zhang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ailing Zhong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yiman Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiajia Du
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qi Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianan Zheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingyao Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Siqi Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Feifei Na
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenghao Lu
- Chengdu OrganoidMed Medical Laboratory, West China Health Valley, Chengdu, Sichuan 610041, China
| | - Jiaming Liu
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaonan Zheng
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lu Yang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Peng Zhang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ping Han
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan 610041, China
| | - Qian Zhong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510000, China
| | - Kai Xiao
- Laboratory of Non-Human Primate Disease Model Research, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanshuo Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yinglan Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hubing Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianghong Man
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing 100850, China
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chengjian Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lunzhi Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhihong Xue
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lu Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Musheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510000, China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yu Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Chong Chen
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
35
|
Ge L, Liu SF. Lentivirus-Mediated Short Hairpin RNA for Follistatin Downregulation Suppresses Tumor Progression in Hypopharyngeal Carcinoma. Curr Med Sci 2022; 42:832-840. [DOI: 10.1007/s11596-022-2615-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/21/2021] [Indexed: 11/24/2022]
|
36
|
Saeed N, Mahjabeen I, Hakim F, Hussain MZ, Mehmood A, Nisar A, Ahmed MW, Kayani MA. Role of Chk1 gene in molecular classification and prognosis of gastric cancer using immunohistochemistry and LORD-Q assay. Future Oncol 2022; 18:2827-2841. [PMID: 35762179 DOI: 10.2217/fon-2021-1546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Purpose: The aim of the current study was to assess the prognostic value of the Chk1 gene in the DNA damage response pathway in gastric cancer (GC). Methods: Expression levels of the Chk1 were measured in 220 GC tumor tissues and adjacent healthy/noncancerous tissues using real-time PCR and immunohistochemical staining. Genomic instability in GC patients was measured using the long-run real-time PCR technique for DNA-damage quantification assay and comet assay. Results: Significantly downregulated expression of Chk1 was observed at the mRNA level (p < 0.0001) and protein level (p < 0.0001). Significantly increased frequency of lesions/10 kb and comets was observed in tumor tissues compared with control tissues. Conclusion: The data suggest that downregulated expression of Chk1 and positive Heliobacter pylori infection status may have prognostic significance in GC.
Collapse
Affiliation(s)
- Nadia Saeed
- Cancer genetics and epigenetic lab, Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Ishrat Mahjabeen
- Cancer genetics and epigenetic lab, Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Farzana Hakim
- Department of Biochemistry, Foundation University Medical College, Islamabad, Pakistan
| | | | - Azhar Mehmood
- Cancer genetics and epigenetic lab, Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Asif Nisar
- Cancer genetics and epigenetic lab, Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Malik Waqar Ahmed
- Cancer genetics and epigenetic lab, Department of Biosciences, COMSATS University, Islamabad, Pakistan.,Pakistan Institute of Rehabilitation Sciences (PIRS), Isra University Islamabad Campus, Islamabad, Pakistan
| | - Mahmood Akhtar Kayani
- Cancer genetics and epigenetic lab, Department of Biosciences, COMSATS University, Islamabad, Pakistan
| |
Collapse
|
37
|
Identification of Five m6A-Related lncRNA Genes as Prognostic Markers for Endometrial Cancer Based on TCGA Database. J Immunol Res 2022; 2022:2547029. [PMID: 35571565 PMCID: PMC9095403 DOI: 10.1155/2022/2547029] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/11/2022] [Indexed: 12/11/2022] Open
Abstract
Background. Due to difficulties involved in its early diagnosis and adequate prognostication, uterine corpus endometrial carcinoma (UCEC) is one of the most serious threats to human health, with the five-year survival rate being as low as roughly 60%. The discovery of specific biomarkers that serve as prognosticators of UCEC is of great significance. The role of N6-methyladenosine- (m6A-) related long noncoding RNAs (lncRNAs) in the pathogenesis of UCEC remains undefined. In this study, we explored the expression profiles of m6A-related lncRNAs of patients with UCEC and identified novel prognostic markers for UCEC. Methods. Gene expression and clinical data were extracted from The Cancer Genome Atlas. Coexpression analysis was performed to identify m6A-related lncRNAs, which were entered into univariate Cox regression models for evaluating the prognosis of UCEC. Clusters of UCEC patients and enrichment pathways were identified using consistent data clustering and gene set enrichment analysis (GSEA). A risk score model was established, and Kaplan–Meier analysis was conducted for investigating overall survival (OS) across two patient groups (high risk and low risk). Lastly, the relationship between the risk score and the cell content of 22 types of immune cells, clusters, age, programmed cell death 1 ligand-1 (PD-L1) expression level, immune score, and pathological grade was analyzed. Results. We identified a total of 2084 lncRNAs associated with m6A, of which 32 lncRNAs were prognostically relevant. Two clusters (clusters 1 and 2) of patients with UCEC were defined; patients in cluster 1 were found to have significantly higher pathological grades and shorter overall survival time compared to those in cluster 2. GSEA showed that “MITOTIC SPINDLE and other pathways” were more enriched in cluster 1. Five major lncRNAs associated with m6A were screened out, and risk score modeling was used for UCEC prognosis prediction. High risk scores were associated with a shorter OS. The risk score was also verified as an independent prognostic indicator for UCEC and was related to immune cell infiltration levels. Finally, we observed a higher pathological grade and greater levels of PD-L1 in the high-risk group than in the low-risk group of patients. Conclusions. m6A-related lncRNAs play an important role in UCEC progression. The risk-based model constructed from the five key m6A-related lncRNAs was implicated in immune cell infiltration and can potentially be an accurate prognosticator for UCEC.
Collapse
|
38
|
Ma A, Biersack B, Goehringer N, Nitzsche B, Höpfner M. Novel Thienyl-Based Tyrosine Kinase Inhibitors for the Treatment of Hepatocellular Carcinoma. J Pers Med 2022; 12:jpm12050738. [PMID: 35629160 PMCID: PMC9146161 DOI: 10.3390/jpm12050738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
New medical treatments are urgently needed for advanced hepatocellular carcinoma (HCC). Recently, we showed the anticancer effects of novel thiophene-based kinase inhibitors. In this study, we further characterized the antineoplastic effects and modes of action of the two most promising inhibitors, Thio-Iva and Thio-Dam, and compared their effects with the clinically relevant multi-kinase inhibitor, sorafenib, in HCC cells. Crystal violet staining and real-time cell growth monitoring showed pronounced antiproliferative effects in Huh-7 and SNU-449 cells with IC50 values in the (sub-)micromolar range. Long-term incubation experiments revealed the reduced clonogenicity of Thio-Iva and Thio-Dam-treated HCC cells. LDH-release tests excluded cytotoxicity as an unspecific mode of action of the inhibitors, while flow cytometry analysis revealed a dose-dependent and pronounced G2/M phase cell cycle arrest and cyclin B1 suppression. Additionally, mitochondria-driven apoptosis was observed through the cytosolic increase of reactive oxygen species, a concomitant PARP cleavage, and caspase-3 induction. Both compounds were found to effectively inhibit the capillary tube formation of endothelial EA.hy926 cells in vitro, pointing towards additional antiangiogenic effects. Antiangiogenic and antineoplastic effects were confirmed in vivo by CAM assays. In summary, the thienyl-acrylonitrile derivatives, Thio-Iva and Thio-Dam, exert significant antineoplastic and antiangiogenic effects in HCC cells.
Collapse
Affiliation(s)
- Andi Ma
- Institute of Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany; (A.M.); (N.G.); (M.H.)
| | - Bernhard Biersack
- Organic Chemistry 1, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany;
| | - Nils Goehringer
- Institute of Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany; (A.M.); (N.G.); (M.H.)
| | - Bianca Nitzsche
- Institute of Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany; (A.M.); (N.G.); (M.H.)
- Correspondence:
| | - Michael Höpfner
- Institute of Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany; (A.M.); (N.G.); (M.H.)
| |
Collapse
|
39
|
Farrukh UB, Bilal A, Zahid H, Iqbal M, Manzoor S, Firdous F, Furqan M, Azeem M, Emwas A, Alazmi M, Gao X, Saleem RSZ, Faisal A. Synthesis and Evaluation of Novel Carboxamides Capable of Causing Centrosome Declustering and Apoptosis in Breast Cancer Cells. ChemistrySelect 2022. [DOI: 10.1002/slct.202104218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Usama B. Farrukh
- Department of Chemistry and Chemical Engineering Syed Babar Ali School of Science and Engineering Lahore University of Management Sciences Lahore 54792 Pakistan
| | - Aishah Bilal
- Department of Biology Syed Babar Ali School of Science and Engineering Lahore University of Management Sciences Lahore 54792 Pakistan
| | - Huda Zahid
- Department of Chemistry and Chemical Engineering Syed Babar Ali School of Science and Engineering Lahore University of Management Sciences Lahore 54792 Pakistan
| | - Maheen Iqbal
- Department of Biology Syed Babar Ali School of Science and Engineering Lahore University of Management Sciences Lahore 54792 Pakistan
| | - Safia Manzoor
- Department of Chemistry and Chemical Engineering Syed Babar Ali School of Science and Engineering Lahore University of Management Sciences Lahore 54792 Pakistan
| | - Farhat Firdous
- Department of Chemistry and Chemical Engineering Syed Babar Ali School of Science and Engineering Lahore University of Management Sciences Lahore 54792 Pakistan
| | - Muhammad Furqan
- Department of Biology Syed Babar Ali School of Science and Engineering Lahore University of Management Sciences Lahore 54792 Pakistan
| | - Muhammad Azeem
- Department of Biology Syed Babar Ali School of Science and Engineering Lahore University of Management Sciences Lahore 54792 Pakistan
| | - Abdul‐Hamid Emwas
- Imaging and Characterization Core Lab King Abdullah University of Science and Technology Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Meshari Alazmi
- Computer, Electrical and Mathematical Sciences and Engineering Division King Abdullah University of Science and Technology Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Xin Gao
- Computer, Electrical and Mathematical Sciences and Engineering Division King Abdullah University of Science and Technology Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Rahman S. Z. Saleem
- Department of Chemistry and Chemical Engineering Syed Babar Ali School of Science and Engineering Lahore University of Management Sciences Lahore 54792 Pakistan
| | - Amir Faisal
- Department of Biology Syed Babar Ali School of Science and Engineering Lahore University of Management Sciences Lahore 54792 Pakistan
| |
Collapse
|
40
|
Second-Generation JK-206 Targets the Oncogenic Signal Mediator RHOA in Gastric Cancer. Cancers (Basel) 2022; 14:cancers14071604. [PMID: 35406376 PMCID: PMC8997135 DOI: 10.3390/cancers14071604] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/14/2022] [Accepted: 03/20/2022] [Indexed: 02/05/2023] Open
Abstract
Ras homologous A (RHOA), a signal mediator and a GTPase, is known to be associated with the progression of gastric cancer (GC), which is the fourth most common cause of death in the world. Previously, we designed pharmacologically optimized inhibitors against RHOA, including JK-136 and JK-139. Based on this previous work, we performed lead optimization and designed novel RHOA inhibitors for greater anti-GC potency. Two of these compounds, JK-206 and JK-312, could successfully inhibit the viability and migration of GC cell lines. Furthermore, using transcriptomic analysis of GC cells treated with JK-206, we revealed that the inhibition of RHOA might be associated with the inhibition of the mitogenic pathway. Therefore, JK-206 treatment for RHOA inhibition may be a new therapeutic strategy for regulating GC proliferation and migration.
Collapse
|
41
|
Ohba S, Tang Y, Johannessen TCA, Mukherjee J. PKM2 Interacts With the Cdk1-CyclinB Complex to Facilitate Cell Cycle Progression in Gliomas. Front Oncol 2022; 12:844861. [PMID: 35392228 PMCID: PMC8981990 DOI: 10.3389/fonc.2022.844861] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/24/2022] [Indexed: 11/13/2022] Open
Abstract
PKM2 is a phosphotyrosine-binding glycolytic enzyme upregulated in many cancers, including glioma, and contributes to tumor growth by regulating cell cycle progression. We noted, however, that in multiple glioma cell lines, PKM2 knock-down resulted in an accumulation of cells in G2-M phase. Moreover, PKM2 knock-down decreased Cdk1 activity while introducing a constitutively active Cdk1 reversed the effects of PKM2 knock-down on cell cycle progression. The means by which PKM2 increases Cdk1 activity have not been described. Transient interaction of T14/Y15-phosphorylated Cdk1 with cyclin B allows Cdk7-mediated pT161 Cdk1 phosphorylation followed by cdc25C-mediated removal of pT14/Y15 and activation of Cdk1 in cycling cells. In the present course of investigation, PKM2 modulation did not influence Cdk7 activity, but phosphotyrosine binding forms of PKM2 co-immunoprecipitated with pY15-containing Cdk1-cyclinB and enhanced formation of active pT161 Cdk1-cyclin B complexes. Moreover, exogenous expression of phosphotyrosine binding forms of PKM2 reversed the effects of PKM2 knock-down on G2-M arrest. We here show that PKM2 binds and stabilize otherwise transient pY15-containing Cdk1-cyclinB complexes that in turn facilitate Cdk1-cyclin B activation and entry of cells into mitosis. These results, therefore, establish metabolic enzyme PKM2 as a direct interactor and activator of Cdk1-cyclin B complex and thereby directly controls mitotic progression and the growth of brain tumor cells.
Collapse
Affiliation(s)
- Shigeo Ohba
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| | - Yongjian Tang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Tor-Christian Aase Johannessen
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
- The Kristian Gerhard Jebsen Brain Tumor Research Centre, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Joydeep Mukherjee
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
- *Correspondence: Joydeep Mukherjee,
| |
Collapse
|
42
|
Wang X, Zhang H, Wang Y, Wang Y, Han Q, Yan H, Yang T, Guo Z. Platinum Complexes as Inhibitors of DNA Repair Protein Ku70 and Topoisomerase IIα in Cancer Cells. Dalton Trans 2022; 51:3188-3197. [DOI: 10.1039/d1dt03700e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Ku70 protein and topoisomerase IIα (Topo IIα) are promising targets of anticancer drugs, which play critical roles in DNA repair and replication processes. Three platinum(II) complexes, [PtCl(NH3)2(9-(pyridin-2-ylmethyl)-9H-carbazole)]NO3 (OPPC), [PtCl(NH3)2(9-(pyridin-3-ylmethyl)-9H-carbazole)]NO3 (MPPC),...
Collapse
|
43
|
Wang QQ, Yin G, Huang JR, Xi SJ, Qian F, Lee RX, Peng XC, Tang FR. Ionizing Radiation-Induced Brain Cell Aging and the Potential Underlying Molecular Mechanisms. Cells 2021; 10:3570. [PMID: 34944078 PMCID: PMC8700624 DOI: 10.3390/cells10123570] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 01/10/2023] Open
Abstract
Population aging is occurring rapidly worldwide, challenging the global economy and healthcare services. Brain aging is a significant contributor to various age-related neurological and neuropsychological disorders, including Alzheimer's disease and Parkinson's disease. Several extrinsic factors, such as exposure to ionizing radiation, can accelerate senescence. Multiple human and animal studies have reported that exposure to ionizing radiation can have varied effects on organ aging and lead to the prolongation or shortening of life span depending on the radiation dose or dose rate. This paper reviews the effects of radiation on the aging of different types of brain cells, including neurons, microglia, astrocytes, and cerebral endothelial cells. Further, the relevant molecular mechanisms are discussed. Overall, this review highlights how radiation-induced senescence in different cell types may lead to brain aging, which could result in the development of various neurological and neuropsychological disorders. Therefore, treatment targeting radiation-induced oxidative stress and neuroinflammation may prevent radiation-induced brain aging and the neurological and neuropsychological disorders it may cause.
Collapse
Affiliation(s)
- Qin-Qi Wang
- Laboratory of Oncology, Center for Molecular Medicine, Health Science Center, School of Basic Medicine, Yangtze University, Jingzhou 434023, China; (Q.-Q.W.); (S.-J.X.)
- Health Science Center, Department of Pathophysiology, School of Basic Medicine, Yangtze University, Jingzhou 434023, China
| | - Gang Yin
- Department of Neurology, Jingzhou Central Hospital, Jingzhou 434023, China;
| | - Jiang-Rong Huang
- Health Science Center, Department of Integrative Medicine, School of Health Sciences, Yangtze University, Jingzhou 434023, China;
| | - Shi-Jun Xi
- Laboratory of Oncology, Center for Molecular Medicine, Health Science Center, School of Basic Medicine, Yangtze University, Jingzhou 434023, China; (Q.-Q.W.); (S.-J.X.)
- Health Science Center, Department of Pathophysiology, School of Basic Medicine, Yangtze University, Jingzhou 434023, China
| | - Feng Qian
- Health Science Center, Department of Physiology, School of Basic Medicine, Yangtze University, Jingzhou 434023, China;
| | - Rui-Xue Lee
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore;
| | - Xiao-Chun Peng
- Laboratory of Oncology, Center for Molecular Medicine, Health Science Center, School of Basic Medicine, Yangtze University, Jingzhou 434023, China; (Q.-Q.W.); (S.-J.X.)
- Health Science Center, Department of Pathophysiology, School of Basic Medicine, Yangtze University, Jingzhou 434023, China
| | - Feng-Ru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore;
| |
Collapse
|
44
|
Zhu XL, Li Q, Shen J, Shan L, Zuo ED, Cheng X. Use of 6 m6A-relevant lncRNA genes as prognostic markers of primary liver hepatocellular carcinoma based on TCGA database. Transl Cancer Res 2021; 10:5337-5351. [PMID: 35116381 PMCID: PMC8797289 DOI: 10.21037/tcr-21-2440] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/23/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is diagnosed at the middle and advanced stages, negating radical treatment. Identifying specific and effective prognostic HCC biomarkers is important and can facilitate the discovery of potential therapeutic targets. N6-methyladenosine (m6A) and long non-coding RNAs (lncRNAs) are associated with the development of multiple tumors. The role of m6A-relevant lncRNAs in the initiation and progression of HCC is unclear. The aim of the present study was to investigate the expression of m6A-relevant lncRNAs in HCC and to identify new prognostic markers of the disease. METHODS Gene expression and clinical data were retrieved from The Cancer Genome Atlas database. m6A-relevant lncRNAs were identified by co-expression analysis and were screened by univariate Cox regression analysis. Different HCC patient clusters were established via consensus clustering. Gene set enrichment analysis (GSEA) was used to determine the cluster enrichment pathways. A risk score model was constructed, and Kaplan-Meier analysis of the overall survival (OS) between cluster 1 (high risk) and cluster 2 (low risk) was performed. Relationships between the clusters, risk scores, and clinicopathological characteristics were clarified. RESULTS Of the 1,852 m6A-relevant lncRNAs identified, 68 had prognostic relevance. The pathological grade, American Joint Committee on Cancer stage, and T stage of cluster 1 were significantly more advanced than those of cluster 2. Based on GSEA, mitotic spindle, G2M_CHECKPOINT, glycolysis, the phosphoinositide 3-kinase (PI3K) protein kinase B (AKT) mammalian target of rapamycin (mTOR) pathway, and DNA repair were more enriched in cluster 1. Six key m6A-relevant lncRNAs were selected to build a risk score model predicting the prognosis of HCC. The OS of patients in the high-risk group was shorter than that of patients in the low-risk group. Risk score was an independent prognostic factor of HCC patients. CONCLUSIONS The findings indicated that m6A-relevant lncRNAs may be important in the progression of HCC. The risk score model based on the 6 key m6A-relevant lncRNAs can accurately predict the prognosis of patients with HCC.
Collapse
Affiliation(s)
- Xiao-Li Zhu
- Department of Hematology and Oncology, Soochow University Affiliated Taicang Hospital (The First People’s Hospital of Taicang), Taicang, China
| | - Qing Li
- Department of Gastroenterology, Soochow University Affiliated Taicang Hospital (The First People’s Hospital of Taicang), Taicang, China
| | - Jie Shen
- Department of Administrative Office, Jiangsu University Affiliated Kunshan Hospital (The First People’s Hospital of Kunshan), Kunshan, China
| | - Li Shan
- Department of Hematology and Oncology, Soochow University Affiliated Taicang Hospital (The First People’s Hospital of Taicang), Taicang, China
| | - Er-Dong Zuo
- Department of Hematology and Oncology, Soochow University Affiliated Taicang Hospital (The First People’s Hospital of Taicang), Taicang, China
| | - Xu Cheng
- Department of Hematology and Oncology, Soochow University Affiliated Taicang Hospital (The First People’s Hospital of Taicang), Taicang, China
| |
Collapse
|
45
|
Cai S, Feng Y, Ye J, Deng Y, Cai Z, Zhu X, Liu R, Zhang Y, Zou Z, Tang Z, Han Z, Hon CT, Zhong W, He H. The prognostic roles of CYP19A1 expression in bladder cancer patients of different genders. Transl Androl Urol 2021; 10:3579-3590. [PMID: 34733654 PMCID: PMC8511542 DOI: 10.21037/tau-21-400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/02/2021] [Indexed: 12/24/2022] Open
Abstract
Background The incidence of bladder cancer (BCa) in male is approximately three to four times higher than in female, but the oncological outcomes in female patients with BCa are significantly worse than in male patients. Although many biomarkers have been identified in recent decades to predict the prognosis of BCa patients, few of them are able to distinguish the prognosis of BCa patients with gender difference. Aromatase encoded by the CYP19A1 gene catalyzes the conversion of androgens to estrogens. In this study, we investigate the prognosis significance of CYP19A1 expression considering the gender difference in BCa patients from four available public databases. Methods Four available public databases of BCa, including GSE13507, TCGA-BLCA, E-MTAB-4321, and E-MTAB-1803, were utilized in this analysis. The overall survival (OS) and progression-free survival (PFS) in different stages and genders were evaluated using the Kaplan-Meier analysis based on the optimal cut-off values of CYP19A1 expression. Then, Gene Set Enrichment Analysis (GSEA) were further performed to explore the potential biologic pathways by altering CYP19A1 expression in BCa patients. Results The results showed that patients with high CYP19A1 expression had a poorer outcome compared with those with low expression in both BCa cohorts in general. Higher CYP19A1 expression in male patients were significantly associated with shorter survival for either non-muscle-invasive bladder cancer (NMIBC) or muscle-invasive bladder cancer (MIBC). However, female NMIBC patients with high CYP19A1 expression were identified to have a better prognosis, whereas high CYP19A1 expression in female MIBC patients were significantly associated with poorer survival. The result of the GSEA showed that different outcomes in female and male patients with NMIBC were related to the interaction of CYP19A1 and the cell-cycle-related pathways. Conclusions These findings demonstrated that CYP19A1 expression might have a potential role in distinguishing the prognosis of female BCa patients dependent on tumor stage. Our results provide new insights for aromatase-mediated BCa therapy.
Collapse
Affiliation(s)
- Shanghua Cai
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yuanfa Feng
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Jianheng Ye
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yulin Deng
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Zhiduan Cai
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuejin Zhu
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Ren Liu
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yixun Zhang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhihao Zou
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhenfeng Tang
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Zhaodong Han
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chi Tin Hon
- Macau Institute of Systems Engineering, Macau University of Science and Technology, Macau, China
| | - Weide Zhong
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.,Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.,Department of Urology, Huizhou Municipal Central Hospital, Huizhou, China.,Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Huichan He
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
46
|
Vakili-Samiani S, Turki Jalil A, Abdelbasset WK, Yumashev AV, Karpisheh V, Jalali P, Adibfar S, Ahmadi M, Hosseinpour Feizi AA, Jadidi-Niaragh F. Targeting Wee1 kinase as a therapeutic approach in Hematological Malignancies. DNA Repair (Amst) 2021; 107:103203. [PMID: 34390915 DOI: 10.1016/j.dnarep.2021.103203] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/26/2021] [Accepted: 08/02/2021] [Indexed: 01/30/2023]
Abstract
Hematologic malignancies include various diseases that develop from hematopoietic stem cells of bone marrow or lymphatic organs. Currently, conventional DNA-damage-based chemotherapy drugs are approved as standard therapeutic regimens for these malignancies. Although many improvements have been made, patients with relapsed or refractory hematological malignancies have a poor prognosis. Therefore, novel and practical therapeutic approaches are required for the treatment of these diseases. Interestingly several studies have shown that targeting Wee1 kinase in the Hematological malignancies, including AML, ALL, CML, CLL, DLBCL, BL, MCL, etc., can be an effective therapeutic strategy. It plays an essential role in regulating the cell cycle process by abrogating the G2-M cell-cycle checkpoint, which provides time for DNA damage repair before mitotic entry. Consistently, Wee1 overexpression is observed in various Hematological malignancies. Also, in healthy normal cells, repairing DNA damages occurs due to G1-S checkpoint function; however, in the cancer cells, which have an impaired G1-S checkpoint, the damaged DNA repair process depends on the G2-M checkpoint function. Thus, Wee1 inhibition could be a promising target in the presence of DNA damage in order to potentiate multiple therapeutic drugs. This review summarized the potentials and challenges of Wee1 inhibition combined with other therapies as a novel effective therapeutic strategy in Hematological malignancies.
Collapse
Affiliation(s)
- Sajjad Vakili-Samiani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia; Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | | | - Vahid Karpisheh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pooya Jalali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Adibfar
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
47
|
Moore KN, Hong DS, Patel MR, Pant S, Ulahannan SV, Jones S, Meric-Bernstam F, Wang JS, Aljumaily R, Hamilton EP, Wittchen ES, Wang X, Lin AB, Bendell JC. A Phase 1b Trial of Prexasertib in Combination with Standard-of-Care Agents in Advanced or Metastatic Cancer. Target Oncol 2021; 16:569-589. [PMID: 34559360 DOI: 10.1007/s11523-021-00835-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND The checkpoint kinase 1 (CHK1) inhibitor prexasertib exhibited modest monotherapy antitumor activity in prior trials, suggesting that combination with chemotherapy or other targeted agents may be needed to maximize efficacy. OBJECTIVES The aim of this study was to determine the recommended phase II dose and schedule of prexasertib in combination with either cisplatin, cetuximab, pemetrexed, or 5-fluorouracil in patients with advanced and/or metastatic cancer, and to summarize preliminary antitumor activity of these combinations. PATIENTS AND METHODS This phase Ib, nonrandomized, open-label study comprised dose-escalation phase(s) with multiple sub-arms evaluating different prexasertib-drug combinations: Part A, prexasertib + cisplatin (n = 63); Part B, prexasertib + cetuximab (n = 41); Part C, prexasertib + pemetrexed (n = 3); Part D, prexasertib + 5-fluorouracil (n =8). Alternate dose schedules/regimens intended to mitigate toxicity and maximize dose exposure and efficacy were also explored in sub-parts. RESULTS In Part A, the maximum tolerated dose (MTD) of prexasertib in combination with cisplatin (75 mg/m2) was declared at 80 mg/m2, with cisplatin administered on Day 1 and prexasertib on Day 2 of a 21-day cycle. The overall objective response rate (ORR) in Part A was 12.7%, and 28 of 55 evaluable patients (50.9%) had a decrease in target lesions from baseline. The most frequent treatment-related adverse events (AEs) in Part A were hematologic, with the most common being white blood cell count decreased/neutrophil count decreased, experienced by 73.0% (any grade) and 66.7% (grade 3 or higher) of patients. In Part B, an MTD of 70 mg/m2 was established for prexasertib administered in combination with cetuximab (500 mg /m2), both administered on Day 1 of a 14-day cycle. The overall ORR in Part B was 4.9%, and 7 of 31 evaluable patients (22.6%) had decreased target lesions compared with baseline. White blood cell count decreased/neutrophil count decreased was also the most common treatment-related AE (56.1% any grade; 53.7% grade 3 or higher). In Parts A and B, hematologic toxicities, even with the addition of prophylactic granulocyte colony-stimulating factor, resulted in frequent dose adjustments (> 60% of patients). In Part C, evaluation of prexasertib + pemetrexed was halted due to dose-limiting toxicities in two of the first three patients; MTD was not established. In Part D, the MTD of prexasertib in combination with 5-fluorouracil (label dose) was declared at 40 mg /m2, both administered on Day 1 of a 14-day cycle. In Part D, overall ORR was 12.5%. CONCLUSIONS This study demonstrated the proof-of-concept that prexasertib can be combined with cisplatin, cetuximab, and 5-fluorouracil. Schedule was a key determinant of the tolerability and feasibility of combining prexasertib with these standard-of-care agents. Reversible hematologic toxicity was the most frequent AE and was dose-limiting. Insights gleaned from this study will inform future combination strategies for the development of prexasertib and next-generation CHK1 inhibitors. CLINICALTRIALS. GOV IDENTIFIER NCT02124148 (date of registration 28 April 2014).
Collapse
Affiliation(s)
- Kathleen N Moore
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 920 Stanton L Young Blvd WP2350, Oklahoma City, OK, 73104, USA. .,Sarah Cannon Research Institute, Nashville, TN, USA.
| | - David S Hong
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Manish R Patel
- Sarah Cannon Research Institute, Nashville, TN, USA.,Florida Cancer Specialists and Research Institute, Sarasota, FL, USA
| | - Shubham Pant
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 920 Stanton L Young Blvd WP2350, Oklahoma City, OK, 73104, USA.,The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Susanna V Ulahannan
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 920 Stanton L Young Blvd WP2350, Oklahoma City, OK, 73104, USA.,Sarah Cannon Research Institute, Nashville, TN, USA
| | | | | | - Judy S Wang
- Sarah Cannon Research Institute, Nashville, TN, USA.,Florida Cancer Specialists and Research Institute, Sarasota, FL, USA
| | - Raid Aljumaily
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 920 Stanton L Young Blvd WP2350, Oklahoma City, OK, 73104, USA.,Sarah Cannon Research Institute, Nashville, TN, USA
| | - Erika P Hamilton
- Sarah Cannon Research Institute, Nashville, TN, USA.,Tennessee Oncology, Nashville, TN, USA
| | | | | | | | - Johanna C Bendell
- Sarah Cannon Research Institute, Nashville, TN, USA.,Tennessee Oncology, Nashville, TN, USA
| |
Collapse
|
48
|
Huang PQ, Boren BC, Hegde SG, Liu H, Unni AK, Abraham S, Hopkins CD, Paliwal S, Samatar AA, Li J, Bunker KD. Discovery of ZN-c3, a Highly Potent and Selective Wee1 Inhibitor Undergoing Evaluation in Clinical Trials for the Treatment of Cancer. J Med Chem 2021; 64:13004-13024. [PMID: 34423975 DOI: 10.1021/acs.jmedchem.1c01121] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Wee1 inhibition has received great attention in the past decade as a promising therapy for cancer treatment. Therefore, a potent and selective Wee1 inhibitor is highly desirable. Our efforts to make safer and more efficacious Wee1 inhibitors led to the discovery of compound 16, a highly selective Wee1 inhibitor with balanced potency, ADME, and pharmacokinetic properties. The chiral ethyl moiety of compound 16 provided an unexpected improvement of Wee1 potency. Compound 16, known as ZN-c3, showed excellent in vivo efficacy and is currently being evaluated in phase 2 clinical trials.
Collapse
Affiliation(s)
- Peter Q Huang
- Zentalis Pharmaceuticals, San Diego, California 92121, United States
| | - Brant C Boren
- Zentalis Pharmaceuticals, San Diego, California 92121, United States
| | - Sayee G Hegde
- Zentalis Pharmaceuticals, San Diego, California 92121, United States
| | - Hui Liu
- Zentalis Pharmaceuticals, San Diego, California 92121, United States
| | - Aditya K Unni
- Zentalis Pharmaceuticals, San Diego, California 92121, United States
| | - Sunny Abraham
- Zentalis Pharmaceuticals, San Diego, California 92121, United States
| | - Chad D Hopkins
- Zentalis Pharmaceuticals, San Diego, California 92121, United States
| | - Sunil Paliwal
- Zentalis Pharmaceuticals, San Diego, California 92121, United States
| | - Ahmed A Samatar
- Zentalis Pharmaceuticals, San Diego, California 92121, United States
| | - Jiali Li
- Zentalis Pharmaceuticals, San Diego, California 92121, United States
| | - Kevin D Bunker
- Zentalis Pharmaceuticals, San Diego, California 92121, United States
| |
Collapse
|
49
|
Cash T, Fox E, Liu X, Minard CG, Reid JM, Scheck AC, Weigel BJ, Wetmore C. A phase 1 study of prexasertib (LY2606368), a CHK1/2 inhibitor, in pediatric patients with recurrent or refractory solid tumors, including CNS tumors: A report from the Children's Oncology Group Pediatric Early Phase Clinical Trials Network (ADVL1515). Pediatr Blood Cancer 2021; 68:e29065. [PMID: 33881209 PMCID: PMC9090141 DOI: 10.1002/pbc.29065] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/06/2021] [Accepted: 04/02/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND Prexasertib (LY2606368) is a novel, second-generation, selective dual inhibitor of checkpoint kinase proteins 1 (CHK1) and 2 (CHK2). We conducted a phase 1 trial of prexasertib to estimate the maximum-tolerated dose (MTD) and/or recommended phase 2 dose (RP2D), to define and describe the toxicities, and to characterize the pharmacokinetics (PK) of prexasertib in pediatric patients with recurrent or refractory solid and central nervous system (CNS) tumors. METHODS Prexasertib was administered intravenously (i.v.) on days 1 and 15 of a 28-day cycle. Four dose levels, 80, 100, 125, and 150 mg/m2 , were evaluated using a rolling-six design. PK analysis was performed during cycle 1. Tumor tissue was examined for biomarkers (CHK1 and TP53) of prexasertib activity. RESULTS Thirty patients were enrolled; 25 were evaluable. The median age was 9.5 years (range: 2-20) and 21 (70%) were male. Twelve patients (40%) had solid tumors and 18 patients (60%) had CNS tumors. There were no cycle 1 or later dose-limiting toxicities. Common cycle 1, drug-related grade 3/4 toxicities (> 10% of patients) included neutropenia (100%), leukopenia (68%), thrombocytopenia (24%), lymphopenia (24%), and anemia (12%). There were no objective responses; best overall response was stable disease in three patients for five cycles (hepatocellular carcinoma), three cycles (ependymoma), and five cycles (undifferentiated sarcoma). The PK appeared dose proportional across the 80-150 mg/m2 dose range. CONCLUSIONS Although the MTD of prexasertib was not defined by this study, 150 mg/m2 administered i.v. on days 1 and 15 of a 28-day cycle was determined to be the RP2D.
Collapse
Affiliation(s)
- Thomas Cash
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Emory University, Atlanta, GA, USA
| | - Elizabeth Fox
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Xiaowei Liu
- Children’s Oncology Group, Monrovia, CA, USA
| | - Charles G. Minard
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX
| | | | - Adrienne C. Scheck
- Center for Cancer and Blood Disorders, Phoenix Children’s Hospital, Institute for Molecular Medicine, Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Brenda J. Weigel
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Cynthia Wetmore
- Center for Cancer and Blood Disorders, Phoenix Children’s Hospital, Institute for Molecular Medicine, Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| |
Collapse
|
50
|
Comparable radiation sensitivity in p53 wild-type and p53 deficient tumor cells associated with different cell death modalities. Cell Death Discov 2021; 7:184. [PMID: 34285189 PMCID: PMC8292512 DOI: 10.1038/s41420-021-00570-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 05/05/2021] [Accepted: 06/25/2021] [Indexed: 11/08/2022] Open
Abstract
Studies of radiation interaction with tumor cells often take apoptosis as the desired results. However, mitotic catastrophe and senescence are also promoted by clinically relevant doses of radiation. Furthermore, p53 is a well-known transcription factor that is closely associated with radiosensitivity and radiation-induced cell death. Therefore, we aimed to investigate the involvement of radiosensitivity, cell death modalities and p53 status in response to carbon-ion radiation (CIR) here. Isogenic human colorectal cancer cell lines HCT116 (p53+/+ and p53-/-) were irradiated with high-LET carbon ions. Cell survival was determined by the standard colony-forming assay. 53BP1 foci were visualized to identify the repair kinetics of DNA double-strand breaks (DSBs). Cellular senescence was measured by SA-β-Gal and Ki67 staining. Mitotic catastrophe was determined with DAPI staining. Comparable radiosensitivities of p53+/+ and p53-/- HCT116 colorectal cells induced by CIR were demonstrated, as well as persistent 53BP1 foci indicated DNA repair deficiency in both cell lines. Different degree of premature senescence in isogenic HCT116 colorectal cancer cells suggested that CIR-induced premature senescence was more dependent on p21 but not p53. Sustained upregulation of p21 played multifunctional roles in senescence enhancement and apoptosis inhibition in p53+/+ cells. p21 inhibition further increased radiosensitivity of p53+/+ cells. Complex cell death modalities rather than single cell death were induced in both p53+/+ and p53-/- cells after 5 Gy CIR. Mitotic catastrophe was predominant in p53-/- cells due to inefficient activation of Chk1 and Chk2 phosphorylation in combination with p53 null. Senescence was the major cell death mechanism in p53+/+ cells via p21-dependent pathway. Taken together, p21-mediated premature senescence might be used by tumor cells to escape from CIR-induced cytotoxicity, at least for a time.
Collapse
|