1
|
Weiss K, Agarkova Y, Zwosta A, Hoevel S, Himmelreich AK, Shumanska M, Etich J, Poschmann G, Brachvogel B, Bogeski I, Mielenz D, Riemer J. A fluorescent sensor for real-time monitoring of DPP8/9 reveals crucial roles in immunity and cancer. Life Sci Alliance 2025; 8:e202403076. [PMID: 40355159 PMCID: PMC12069513 DOI: 10.26508/lsa.202403076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 04/30/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025] Open
Abstract
Dipeptidyl peptidases 8 and 9 (DPP8/9) are critical for the quality control of mitochondrial and endoplasmic reticulum protein import, immune regulation, cell adhesion, and cell migration. Dysregulation of DPP8/9 is associated with pathologies including tumorigenesis and inflammation. Commonly, DPP8/9 activity is analysed by in vitro assays using artificial substrates, which allow neither continuously monitoring DPP8/9 activity in individual, living cells nor detecting effects from endogenous interactors and posttranslational modifications. Here, we developed DiPAK (for DPP8/9 activity sensor based on AK2), a ratiometric genetically encoded fluorescent sensor, which enables studying DPP8/9 activity in living cells. Using DiPAK, we determined the dynamic range of DPP8/9 activity in cells overexpressing or lacking DPP9. We identified distinct activity levels among melanoma cell lines and found that LPS-induced primary B-cell activation depends on DPP8/9 as the absence of DPP8/9 activity results in apoptotic but not pyroptotic cell death. Consistently, we observed increasing DPP8/9 activity during B-cell maturation. Overall, DiPAK is a versatile tool for real-time single-cell monitoring of DPP8/9 activity in a broad range of cells and organisms.
Collapse
Affiliation(s)
- Konstantin Weiss
- Redox Metabolism Group, Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Yelizaveta Agarkova
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Alexandra Zwosta
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sarah Hoevel
- Redox Metabolism Group, Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Ann-Kathrin Himmelreich
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Magdalena Shumanska
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Julia Etich
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Gereon Poschmann
- Institute for Molecular Medicine, Proteome Research, University Hospital and Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Bent Brachvogel
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Ivan Bogeski
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Dirk Mielenz
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jan Riemer
- Redox Metabolism Group, Institute for Biochemistry, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
2
|
Oliveira MDS, Dias IRSB, Costa RGA, Rodrigues ACBDC, Silva SLR, Soares MBP, Dias RB, Valverde LF, Gurgel Rocha CA, Batista AA, Correa RS, Silva VR, Granado Pina ET, Bezerra DP. Ru(II)-thymine complex suppresses acute myeloid leukemia stem cells by inhibiting NF-κB signaling. Biomed Pharmacother 2025; 187:118080. [PMID: 40288174 DOI: 10.1016/j.biopha.2025.118080] [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: 10/07/2024] [Revised: 04/02/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
Acute myeloid leukemia (AML) is a lethal hematologic malignancy caused by leukemic blasts that fail to mature normally. AML has a high relapse rate, primarily due to a small subset known as leukemic stem cells (LSCs). In this work, we investigated the ability of a Ru(II)-thymine complex (RTC) with the formula [Ru(PPh3)2(Thy)(bipy)]PF6 (where PPh3 = triphenylphosphine, Thy = thymine, and bipy = 2,2'-bipyridine) to suppress AML LSCs. RTC exhibited potent cytotoxicity toward both solid and hematologic malignancies and suppressed primary AML LSCs, as observed by the reduction in the CD34 +CD38- cell population. In the AML cell line KG-1a, which has an LSC-like population, RTC reduced the number of CD34 + and CD123 + cells. A reduction in leukemic blasts was detected in the bone marrow of RTC-treated NSG mice bearing KG-1a xenografts. Increased DNA fragmentation, YO-PRO-1 staining, active caspase-3 and cleaved PARP (Asp 214) levels, and mitochondrial superoxide levels were detected in RTC-treated KG-1a cells. The pancaspase inhibitor Z-VAD-(OMe)-FMK, but not the antioxidant N-acetylcysteine, partially prevented RTC-induced cell death in KG-1a cells, indicating that RTC induces caspase-mediated apoptosis in KG-1a cells via an oxidative stress-independent pathway. In molecular mechanism studies, transcripts of the NF-κB inhibitor NFKBIA were upregulated, and the level of NF-κB p65 phosphorylated at the Ser529 residue was reduced in RTC-treated KG-1a cells, indicating that RTC may inhibit NF-κB signaling. Overall, these results indicate the anti-AML potential of RTC in AML LSCs via the suppression of NF-κB signaling.
Collapse
Affiliation(s)
- Maiara de S Oliveira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil
| | - Ingrid R S B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil
| | - Rafaela G A Costa
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil
| | | | - Suellen L R Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil; SENAI Institute for Innovation in Advanced Health Systems, SENAI CIMATEC, Salvador, BA 41650-010, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil; Department of Biological Sciences, State University of Feira de Santana, Feira de Santana, BA 44036-900, Brazil
| | - Ludmila F Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil; Department of Dentistry, Federal University of Sergipe, Lagarto, SE 49400-000, Brazil
| | - Clarissa A Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil; Department of Propaedeutics, Faculty of Dentistry, Federal University of Bahia (UFBA), Salvador, BA 40301-155, Brazil
| | - Alzir A Batista
- Department of Chemistry, Federal University of São Carlos, São Carlos, SP 13561-901, Brazil
| | - Rodrigo S Correa
- Department of Chemistry, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - Valdenizia R Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil
| | - Eugênia T Granado Pina
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil; Research Center, National Cancer Institute (INCA), Rio de Janeiro, RJ 20230-130, Brazil
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil.
| |
Collapse
|
3
|
Hou X, Zanfagnin V, Xu C, Jessen E, Liu Y, Wang C, Huang Y, Fontaine SD, Santi DV, Colon-Otero G, Gill SE, Glaser GE, Butler KA, Bakkum-Gamez JN, Dowdy SC, Oberg AL, Larson MC, Atkinson HJ, Duffield LN, Peterson KL, Kaufmann SH, Weroha SJ. Antitumor activity of rucaparib plus PLX038A in serous endometrial carcinoma. J Exp Clin Cancer Res 2025; 44:150. [PMID: 40390012 PMCID: PMC12087071 DOI: 10.1186/s13046-025-03406-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 05/02/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Serous endometrial cancer (SEC) is a genomically and morphologically distinct endometrial cancer (EC) subtype with a poor progression-free and overall survival. The development of novel therapies is needed to improve outcomes. METHODS We used serous and serous-like EC patient-derived xenografts (PDXs) to test a novel drug combination in vitro and in vivo: rucaparib and pegylated SN-38 (PLX038A). Sensitivity to treatment was correlated with indicators of homologous recombination (HR) deficiency. Efficacy in fresh primary patient tumors was also tested ex vivo. RESULTS Five of eight PDXs had genomic instability scores ≥ 42, but only one of these five had evidence of HR deficiency in assays of irradiation-induced RAD51 foci formation. Moreover, PARP inhibitor (PARPi) monotherapy failed to induce regressions in any of the five SEC models treated with rucaparib in vivo, suggesting limited clinical activity of PARPi in SEC. In further studies, we assessed the response of these models to the sustained release topoisomerase 1 inhibitor, PLX038A, as monotherapy and in combination with rucaparib ex vivo and in vivo. Results of these studies showed that PLX038A had limited monotherapy activity, but combination therapy induced significant regressions in two of five SEC PDXs and markedly slowed tumor growth in the other three regardless of underlying homologous recombination repair deficiency. In addition, 11 of 20 (55%) primary tumors showed synergy with rucaparib + SN-38. CONCLUSIONS Collectively, these studies identify a set of genomically characterized PDX models for preclinical testing of potential SEC therapies and a therapeutic combination that warrants further preclinical investigation.
Collapse
Affiliation(s)
- Xiaonan Hou
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Conway Xu
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Mayo Clinic, MN, 55905, USA
| | - Erik Jessen
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yuanhang Liu
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Chen Wang
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yajue Huang
- Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA
| | - Shaun D Fontaine
- ProLynx LLC, 455 Mission Bay Blvd, South San Francisco, CA, 94158, USA
| | - Daniel V Santi
- ProLynx LLC, 455 Mission Bay Blvd, South San Francisco, CA, 94158, USA
| | | | - Sarah E Gill
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Mayo Clinic, MN, 55905, USA
| | - Gretchen E Glaser
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Mayo Clinic, MN, 55905, USA
| | - Kristina A Butler
- Medical & Surgical Gynecology Department, Mayo Clinic, Phoenix, AZ, USA
| | - Jamie N Bakkum-Gamez
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Mayo Clinic, MN, 55905, USA
| | - Sean C Dowdy
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Mayo Clinic, MN, 55905, USA
| | - Ann L Oberg
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Melissa C Larson
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Hunter J Atkinson
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Laura N Duffield
- Division of Oncology Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Kevin L Peterson
- Division of Oncology Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Scott H Kaufmann
- Division of Oncology Research, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - S John Weroha
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Oncology, Mayo Clinic College of Medicine, 200 First St. SW, Rochester, MN, 55905, USA.
| |
Collapse
|
4
|
Nousiainen R, Eloranta K, Saarela J, Hassinen A, Luck TJ, Cairo S, Indersie E, Potdar S, Feodoroff MJ, Lohi J, Paavolainen L, Wilson DB, Pietiäinen V, Heikinheimo M, Pihlajoki M. Functional screening identifies kinesin spindle protein inhibitor filanesib as a potential treatment option for hepatoblastoma. NPJ Precis Oncol 2025; 9:122. [PMID: 40281281 PMCID: PMC12032252 DOI: 10.1038/s41698-025-00915-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Hepatoblastoma is a rare pediatric liver malignancy usually treated with surgery and chemotherapy. To explore new treatment options for hepatoblastoma, drug screening was performed using six cell models established from aggressive hepatoblastoma tumors and healthy pediatric primary hepatocytes. Of the 527 screened compounds, 98 demonstrated cancer-selective activity in at least one hepatoblastoma model. The kinesin spindle protein (KSP) inhibitor filanesib was effective in all models and was further evaluated. Filanesib induced G2/M arrest and apoptosis in hepatoblastoma cells at concentrations tolerable to primary hepatocytes. Prominent nuclear fragmentation was observed in filanesib-treated hepatoblastoma cells. Genes participating in cell cycle regulation were noted to be differentially expressed after filanesib treatment. Filanesib reduced the rate of tumor growth in 4/5 hepatoblastoma mice models. One of these models showed complete growth arrest. Our results suggest that filanesib is a potential candidate for hepatoblastoma treatment and should be investigated in future clinical trials.
Collapse
Affiliation(s)
- Ruth Nousiainen
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Katja Eloranta
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland.
| | - Jani Saarela
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Antti Hassinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Tamara J Luck
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Stefano Cairo
- XenTech, Evry, France
- Champions Oncology, Hackensack, NJ, USA
- Istituto di Ricerca Pediatrica, Padova, Italy
| | | | - Swapnil Potdar
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Michaela J Feodoroff
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Jouko Lohi
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Lassi Paavolainen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - David B Wilson
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Vilja Pietiäinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Markku Heikinheimo
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research, Tampere University, Tampere, Finland
| | - Marjut Pihlajoki
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
5
|
Kachranlouei L, Hosseinzadeh H, Karimi G, Rajabian F, Mehri S. Ameliorative effects of osthole on acrylamide-induced neurotoxicity in PC12 cells: Role of oxidative stress, apoptosis and ERK pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4361-4372. [PMID: 39470816 DOI: 10.1007/s00210-024-03560-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/20/2024] [Indexed: 11/01/2024]
Abstract
The possible protective effects of osthole on acrylamide-induced neurotoxicity in PC12 cells. Cells were pretreated with different concentrations of osthole (1- 25 μM) for 24 h and then the IC50 value of acrylamide (5 mM) was added. After 24 h, cell viability and intracellular ROS content were detected by MTT assay and DCF-DA methods, respectively. Also, DNA fragmentation in apoptotic cells was determined by propidium iodide assay, and apoptosis (Caspase-3, Bax, Bcl-2, ERK, and P-ERK) was measured by the western blot method. Exposing PC12 cells to acrylamide diminished cell viability, and enhanced the intracellular ROS generation and the percentage of apoptotic cells. Furthermore, acrylamide elevated the P-ERK/ERK and Bax/Bcl-2 ratio, and the level of cleaved caspase-3 protein in PC12 cells. Pretreating cells with osthole enhanced cell viability and reduced ROS generation. Also, osthole (10 μM) significantly reduced P-ERK/ERK and Bax/Bcl-2 ratio, the level of cleaved caspase-3 protein, and the percentage of apoptotic cells in comparison to the acrylamide group. Osthole can exhibit a protective effect on the neurotoxicity of acrylamide through the inhibition of oxidative stress and apoptosis in PC12 cells.
Collapse
Affiliation(s)
- Leili Kachranlouei
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, P. O. Box, Mashhad, 1365-91775, IR, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, P. O. Box, Mashhad, 1365-91775, IR, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Rajabian
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soghra Mehri
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, P. O. Box, Mashhad, 1365-91775, IR, Iran.
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Alanazi FS, Alkahtani HM, Abdel-Aziz AAM, El-Azab AS, Asiri HH, Bakheit AH, Al-Omary FA. Synthesis, Antitumor Activities, and Apoptosis-Inducing Activities of Schiff's Bases Incorporating Imidazolidine-2,4-dione Scaffold: Molecular Docking Studies and Enzymatic Inhibition Activities. Pharmaceuticals (Basel) 2025; 18:496. [PMID: 40283934 PMCID: PMC12030650 DOI: 10.3390/ph18040496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objective: Cancer is the leading cause of death worldwide despite the diversity of antitumor therapies, which highlights the necessity to explore new anticancer agents. Methods: We synthesized 5,5-diphenylhydantoin derivatives including Schiff's bases 7-27 and evaluated their cytotoxicity via the MTT assay. Enzymatic inhibition assays, cell cycle and apoptosis analyses, and molecular docking studies were also conducted. Results: Derivative 24 demonstrated the highest cytotoxic activity, with IC50 values of 12.83 ± 0.9 μM, 9.07 ± 0.8 μM, and 4.92 ± 0.3 μM against the cell lines HCT-116, HePG-2, and MCF-7, respectively. Compounds 10, 13, and 21 showed potent antitumor activities versus the examined cell lines (average IC50 = 13.2, 14.5, and 13.1 μM), respectively; moreover, these compounds also demonstrated promising EGFR and HER2 inhibitory activities, with IC50 values in the range 0.28-1.61 µM. Derivative 24 displayed the highest EGFR and HER2 inhibitory activity values (IC50 = 0.07 and 0.04 µM), respectively, which were close to those of the reference drugs erlotinib and lapatinib. Therefore, compound 24 was selected for further examinations and exhibited an inducing effect on apoptosis via diminishing the anti-apoptotic protein levels of BCL-2 (8.598 ± 0.29 ng/mL) and MCL-1 (261.20 ± 8.97 pg/mL) and promoting cell cycle arrest at the G2/M phase (33.46%). The binding relationships between compound 24 and the active sites of EGFR and HER2, which are similar to the co-crystallized inhibitors, were investigated using a molecular docking approach. Conclusions: These findings provide insights into the potential anticancer activities of the synthesized derivatives for further optimization to achieve therapeutic use.
Collapse
Affiliation(s)
- Fhdah S. Alanazi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | | | - Alaa A.-M. Abdel-Aziz
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Adel S. El-Azab
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Hanadi H. Asiri
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | | | | |
Collapse
|
7
|
Cheng Y, Zhang K, Liu J, Liu G. Is orbital adipose tissue obesity-privileged? The relationship between small adipocyte size and metabolically healthy state from the view of orbital fat. J Endocrinol Invest 2025:10.1007/s40618-025-02568-7. [PMID: 40120074 DOI: 10.1007/s40618-025-02568-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/09/2025] [Indexed: 03/25/2025]
Abstract
PURPOSE White adipose tissue (WAT) expands by increasing adipocyte size (hypertrophy) and/or number (hyperplasia) to handle excess energy and plays a key homeostatic role in lipid metabolism. Hypertrophic adipocytes have many impaired biological functions. In contrast, hyperplastic adipocytes can reduce the negative metabolic effects of obesity. Thus, understanding the mechanisms of adaptive WAT expansion is essential for optimizing lipid storage and preventing the adverse metabolic consequences of obesity. Hedgehog (Hh) signaling has been shown to improve adipose health and can be a pharmacological target to ameliorate obesity-induced metabolic abnormalities. Clinically, we found that the size of adipocytes in orbital fat (OF) is less affected by obesity, and we hypothesized that OF possesses a relatively metabolically healthy profile. METHODS To verify our hypothesis, we identified multiple hallmarks of healthy adipose tissue in OF using a combination of bioinformatics-based transcriptomics analyses and experimental methods. RESULTS Our results revealed that compared with abdominal subcutaneous fat (SF), OF had a smaller cell size, more dynamic ability to remodel the adipose extracellular matrix (ECM), higher vascular supply, and less macrophage infiltration. OF also showed promising adipogenic and proliferative capabilities and a healthy adipocytokine secretion pattern. Moreover, the Hh signaling was activated in OF and may influence depot-specific adipose health. CONCLUSION These findings collectively support that OF is generally in a naturally metabolically healthy state with high expandability and obesity-free privilege, providing new therapeutic ideas for obesity-related metabolic dysfunctions.
Collapse
Affiliation(s)
- Y Cheng
- Department of Plastic and Reconstructive Surgery, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - K Zhang
- Department of Plastic and Reconstructive Surgery, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - J Liu
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu, Bengbu, Anhui, China.
| | - G Liu
- Department of Plastic and Reconstructive Surgery, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
| |
Collapse
|
8
|
Ciurus S, Elewa MAF, Palmer MA, Wolf A, Hector M, Fuhrmann DC, Thomas D, Gurke R, Schwalm MP, Berger L, Zech TJ, Burgers LD, Marschalek R, Geisslinger G, Knapp S, Langmann T, Bracher F, Weigert A, Fürst R. Inhibition of DYRK1B BY C81 impedes inflammatory processes in leukocytes by reducing STAT3 activity. Cell Mol Life Sci 2025; 82:85. [PMID: 39985685 PMCID: PMC11846820 DOI: 10.1007/s00018-025-05579-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 10/15/2024] [Accepted: 01/05/2025] [Indexed: 02/24/2025]
Abstract
Chronic inflammatory diseases are a significant global burden and are associated with dysregulated resolution of inflammation. Therefore, promoting the process of resolution is a promising therapeutic approach. This study presents the potent anti-inflammatory and pro-resolving effects of a natural product-derived compound called C81. Administration of C81 in a therapeutic window resolved inflammation in the murine imiquimod-induced psoriasis model, and reduced microglial infiltration in a laser-induced choroidal neovascularisation model. Investigations into the underlying mechanisms of C81 identified the DYRK1B/STAT3 axis as a new regulator of inflammatory processes in leukocytes. The inhibition of DYRK1B by C81 resulted in attenuated STAT3 phosphorylation. The depletion of STAT3-regulated gene expression led to the inhibition of leukocyte adhesion and migration due to reduced integrin activation, and in addition to the inhibition of the release of pro-inflammatory mediators such as cytokines and eicosanoids. Importantly, the pro-resolving effects of C81 included the cell type-specific induction of apoptosis in neutrophils and a subsequent increase in efferocytosis. In conclusion, we report the DYRK1B/STAT3 axis as a novel and promising therapeutic target for activating the resolution of inflammation.
Collapse
Affiliation(s)
- Sarah Ciurus
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - Mohammed A F Elewa
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
- Department of Biochemistry, Faculty of Pharmacy, Kafr El-Sheikh University, Karf El-Sheikh, Egypt
| | - Megan A Palmer
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Anne Wolf
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine, University of Cologne, University Hospital Cologne, Cologne, Germany
- Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Mandy Hector
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine, University of Cologne, University Hospital Cologne, Cologne, Germany
| | - Dominik C Fuhrmann
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Dominique Thomas
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Frankfurt, Germany
| | - Robert Gurke
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Frankfurt, Germany
| | - Martin P Schwalm
- Institute of Pharmaceutical Chemistry and Buchmann Institute Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Lena Berger
- Institute of Pharmaceutical Chemistry and Buchmann Institute Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Thomas J Zech
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
- Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Luisa D Burgers
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Frankfurt, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry and Buchmann Institute Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine, University of Cologne, University Hospital Cologne, Cologne, Germany
- Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Franz Bracher
- Pharmaceutical Chemistry, Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Robert Fürst
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany.
- Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
9
|
Scherer B, Bogun L, Koch A, Jäger P, Maus U, Schmitt L, Krings KS, Wesselborg S, Haas R, Schroeder T, Geyh S. Antineoplastic therapy affects the in vitro phenotype and functionality of healthy human bone marrow-derived mesenchymal stromal cells. Arch Toxicol 2025; 99:393-406. [PMID: 39531065 PMCID: PMC11742341 DOI: 10.1007/s00204-024-03898-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
While antineoplastic therapies aim to specifically target cancer cells, they may also exert adverse effects on healthy tissues, like healthy hematopoietic stem and progenitor cells (HSPC), leading to hematotoxicity as a common side effect. Mesenchymal stromal cells (MSC) are a major component of the bone marrow (BM) microenvironment, regulating normal hematopoiesis, while their susceptibility to anticancer therapies and contribution to therapy-related hematotoxicity remains largely unexplored. To address this, we investigated the effects of etoposide, temozolomide, 5-azacitidine, and venetoclax on healthy BM-derived MSC functionality. Doses below therapeutic effects of etoposide (0.1-0.25 µM) inhibited cellular growth and induced cellular senescence in healthy MSC, accompanied by an increased mRNA expression of CDKN1A, decreased trilineage differentiation capacity, and insufficient hematopoietic support. Pharmacological doses of 5-azacitidine (2.5 µM) shifted MSC differentiation capacity by inhibiting osteogenic capacity but enhancing the chondrogenic lineage, as demonstrated by histochemical staining and on mRNA level. At the highest clinically relevant dose, neither venetoclax (40 nM) nor temozolomide (100 µM) exerted any effects on MSC but clearly inhibited cellular growth of cancer cell lines and primary healthy HSPC, pointing to damage to hematopoietic cells as a major driver of hematotoxicity of these two compounds. Our findings show that besides HSPC, also MSC are sensitive to certain antineoplastic agents, resulting in molecular and functional alterations that may contribute to therapy-related myelosuppression. Understanding these interactions could be helpful for the development of strategies to preserve BM MSC functionality during different kinds of anticancer therapies.
Collapse
Affiliation(s)
- Bo Scherer
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Lucienne Bogun
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Annemarie Koch
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Paul Jäger
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Uwe Maus
- Department of Orthopedic Surgery and Traumatology, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University, Moorenstraße 5, 40225, Duesseldorf, Germany
| | - Laura Schmitt
- Institute for Molecular Medicine 1, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University, Universitätsstraße 1, 40225, Duesseldorf, Germany
| | - Karina S Krings
- Institute for Molecular Medicine 1, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University, Universitätsstraße 1, 40225, Duesseldorf, Germany
| | - Sebastian Wesselborg
- Institute for Molecular Medicine 1, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University, Universitätsstraße 1, 40225, Duesseldorf, Germany
| | - Rainer Haas
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Thomas Schroeder
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany.
| | - Stefanie Geyh
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
10
|
Krings KS, Wassenberg TR, Cea-Medina P, Schmitt L, Lechtenberg I, Llewellyn TR, Qin N, Gohlke H, Wesselborg S, Müller TJJ. Novel 4-alkoxy Meriolin Congeners Potently Induce Apoptosis in Leukemia and Lymphoma Cells. Molecules 2024; 29:6050. [PMID: 39770138 PMCID: PMC11676355 DOI: 10.3390/molecules29246050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/10/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Meriolins (3-(pyrimidin-4-yl)-7-azaindoles) are synthetic hybrids of the naturally occurring alkaloids variolin and meridianin and display a strong cytotoxic potential. We have recently shown that the novel derivative meriolin16 is highly cytotoxic in several lymphoma and leukemia cell lines as well as in primary patient-derived lymphoma and leukemia cells and predominantly targets cyclin-dependent kinases (CDKs). Here, we efficiently synthesized nine novel 2-aminopyridyl meriolin congeners (3a-3i), i.e., pyrimeriolins, using a one-pot Masuda borylation-Suzuki coupling (MBSC) sequence, with eight of them bearing lipophilic alkoxy substituents of varying length, to systematically determine the influence of the alkoxy sidechain length on the biological activity. All the synthesized derivatives displayed a pronounced cytotoxic potential, with six compounds showing IC50 values in the nanomolar range. Derivatives 3b-3f strongly induced apoptosis and activated caspases with rapid kinetics within 3-4 h in Jurkat leukemia and Ramos lymphoma cells. The induction of apoptosis by the most potent derivative 3e was mediated by the intrinsic mitochondrial death pathway, as it was blocked in caspase-9 deficient and Apaf-1 knockdown Jurkat cells. However, as recently shown for meriolin16, derivative 3e was able to induce apoptosis in the Jurkat cells overexpressing the antiapoptotic protein Bcl-2. Since tumor cells often inactivate the intrinsic mitochondrial apoptosis pathway (e.g., by overexpression of Bcl-2), these meriolin congeners represent promising therapeutic agents for overcoming therapeutic resistance.
Collapse
Affiliation(s)
- Karina S. Krings
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany; (K.S.K.); (L.S.); (I.L.)
| | - Tobias R. Wassenberg
- Institute of Organic Chemistry and Macromolecular Chemistry, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany;
| | - Pablo Cea-Medina
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany; (P.C.-M.); (H.G.)
| | - Laura Schmitt
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany; (K.S.K.); (L.S.); (I.L.)
| | - Ilka Lechtenberg
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany; (K.S.K.); (L.S.); (I.L.)
| | - Tanya R. Llewellyn
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, D-40225 Düsseldorf, Germany; (T.R.L.); (N.Q.)
| | - Nan Qin
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, D-40225 Düsseldorf, Germany; (T.R.L.); (N.Q.)
- Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf (CIO ABCD), University Hospital Düsseldorf, Moorenstraße 5, D-40225 Düsseldorf, Germany
| | - Holger Gohlke
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany; (P.C.-M.); (H.G.)
- Institute of Bio- and Geosciences (IBG-4: Bioinformatics), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, D-52425 Jülich, Germany
| | - Sebastian Wesselborg
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany; (K.S.K.); (L.S.); (I.L.)
- Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf (CIO ABCD), University Hospital Düsseldorf, Moorenstraße 5, D-40225 Düsseldorf, Germany
| | - Thomas J. J. Müller
- Institute of Organic Chemistry and Macromolecular Chemistry, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany;
| |
Collapse
|
11
|
Attia MH, Lasheen DS, Samir N, Taher AT, Abdel-Aziz HA, Abou El Ella DA. Design, Synthesis and Molecular Modeling of Pyrazolo[1,5- a]pyrimidine Derivatives as Dual Inhibitors of CDK2 and TRKA Kinases with Antiproliferative Activity. Pharmaceuticals (Basel) 2024; 17:1667. [PMID: 39770509 PMCID: PMC11678221 DOI: 10.3390/ph17121667] [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: 10/31/2024] [Revised: 12/04/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND The increasing prevalence of drug resistance in cancer therapy underscores the urgent need for novel therapeutic approaches. Dual enzyme inhibitors, targeting critical kinases such as CDK2 and TRKA, represent a promising strategy. The goal of this investigation was to design, synthesize, and evaluate a set of pyrazolo[1,5-a]pyrimidine derivatives for their dual inhibition potential toward CDK2 and TRKA kinases, along with their potential antiproliferative against cancer cell lines. METHODS A set of pyrazolo[1,5-a]pyrimidine derivatives (6a-t, 11a-g, and 12) was synthesized and subjected to in vitro enzymatic assays to determine their inhibitory activity against CDK2 and TRKA kinases. Selected compounds were further assessed for antiproliferative effects across the set of 60 cell lines from the NCI, representing various human cancer types. Additionally, simulations of molecular docking were conducted to explore the modes of binding for the whole active compounds and compare them with known inhibitors. RESULTS Compounds 6t and 6s exhibited potent dual inhibitory activity, showing an IC50 = 0.09 µM and 0.23 µM against CDK2, and 0.45 µM against TRKA, respectively. These results were comparable to reference inhibitors ribociclib (CDK2, IC50 = 0.07 µM) and larotrectinib (TRKA, IC50 = 0.07 µM). Among the studied derivatives, compound 6n displayed a notable broad-spectrum anticancer activity, achieving a mean growth inhibition (GI%) of 43.9% across 56 cell lines. Molecular docking simulations revealed that the synthesized compounds adopt modes of binding similar to those of the lead inhibitors. Conclusions: In this study, prepared pyrazolo[1,5-a]pyrimidine derivatives demonstrated significant potential as dual CDK2/TRKA inhibitors, and showed potent anticancer activity toward diverse cancer cell lines. These findings highlight their potential as key compounds for the design of novel anticancer therapeutics.
Collapse
Affiliation(s)
- Mohamed H. Attia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, October 6 University (O6U), Giza 12585, Egypt
| | - Deena S. Lasheen
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt; (D.S.L.); (N.S.)
| | - Nermin Samir
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt; (D.S.L.); (N.S.)
| | - Azza T. Taher
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt;
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, October 6 University (O6U), Giza 12585, Egypt
| | - Hatem A. Abdel-Aziz
- Department of Applied Organic Chemistry, National Research Center, Cairo 12622, Egypt;
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia St., Alexandria 21648, Egypt
| | - Dalal A. Abou El Ella
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt; (D.S.L.); (N.S.)
| |
Collapse
|
12
|
Schmitt L, Krings KS, Wolsing A, Buque X, Zimmermann M, Flores-Romero H, Lenz T, Lechtenberg I, Peter C, Stork B, Teusch N, Proksch P, Stühler K, García-Sáez AJ, Reichert AS, Aspichueta P, Bhatia S, Wesselborg S. Targeting mitochondrial metabolism by the mitotoxin bromoxib in leukemia and lymphoma cells. Cell Commun Signal 2024; 22:541. [PMID: 39533399 PMCID: PMC11558866 DOI: 10.1186/s12964-024-01913-2] [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: 04/10/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Targeting mitochondrial metabolism represents a promising approach for cancer treatment. Here, we investigated the mitotoxic potential of the polybrominated diphenyl ether bromoxib, a natural compound isolated from the marine sponge Dysidea family. We could show that bromoxib comprised strong cytotoxicity in different leukemia and lymphoma cell lines (such as HL60, HPBALL, Jurkat, K562, KOPTK1, MOLT4, SUPB15 and Ramos), but also in solid tumor cell lines (such as glioblastoma cell lines SJ-GBM2 and TP365MG). Bromoxib activated the mitochondrial death pathway as evidenced by the rapid translocation of Bax to the mitochondria and the subsequent mitochondrial release of Smac. Accordingly, bromoxib-induced apoptosis was blocked in caspase 9 deficient Jurkat cells and Jurkat cells overexpressing the antiapoptotic protein Bcl-2. In addition, we could show that bromoxib functioned as an uncoupler of the electron transport chain with similar rapid kinetics as CCCP in terms of dissipation of the mitochondrial membrane potential (ΔΨm), processing of the dynamin-like GTPase OPA1 and subsequent fragmentation of mitochondria. Beyond that, bromoxib strongly abrogated ATP production via glycolysis as well as oxidative phosphorylation (OXPHOS) by targeting electron transport chain complexes II, III, and V (ATP-synthase) in Ramos lymphoma cells. Thus, bromoxib's potential to act on both cytosolic glycolysis and mitochondrial respiration renders it a promising agent for the treatment of leukemia and lymphoma.
Collapse
Affiliation(s)
- Laura Schmitt
- Institute for Molecular Medicine I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Karina S Krings
- Institute for Molecular Medicine I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Andre Wolsing
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Xabier Buque
- Department of Physiology, Faculty of Medicine and Nursing, Universidad del País Vasco, Vitoria-gasteiz, Spain
| | - Marcel Zimmermann
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Hector Flores-Romero
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, 50931, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
- Ikerbasque, Basque Foundation for Science, Bilbao, 48013, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Thomas Lenz
- Molecular Proteomics Laboratory, Biological-Medical-Research Centre (BMFZ), Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Ilka Lechtenberg
- Institute for Molecular Medicine I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Christoph Peter
- Institute for Molecular Medicine I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Björn Stork
- Institute for Molecular Medicine I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Nicole Teusch
- Institute of Pharmaceutical Biology and Biotechnology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Peter Proksch
- Institute of Pharmaceutical Biology and Biotechnology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory, Biological-Medical-Research Centre (BMFZ), Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Ana J García-Sáez
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, 50931, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
| | - Andreas S Reichert
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, Universidad del País Vasco, Vitoria-gasteiz, Spain
- Biobizkia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Sanil Bhatia
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Sebastian Wesselborg
- Institute for Molecular Medicine I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany.
| |
Collapse
|
13
|
Musa M, Bello M, Agwamba EC. Synthesis, Molecular Docking, and Anticancer Screening of Ester-Based Thiazole Derivatives. Chem Biodivers 2024; 21:e202401159. [PMID: 39292150 DOI: 10.1002/cbdv.202401159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/29/2024] [Indexed: 09/19/2024]
Abstract
This study investigates the potential of five compounds as novel anticancer agents. We examined their efficacy, mechanisms of action, and impact on various cancer cell lines, through a comprehensive set of experiments. Notably, compound 3e demonstrated superior activity compared to the positive control cisplatin, with a GI50 value of 6.3±0.7 μM against the breast cancer cell line (MCF-7). Compound 3b also displayed remarkable growth inhibition, yielding GI50 values of 8.7±0.2 μM (MCF-7) and 8.9±0.5 μM against the colon cancer cell line (HCT-116). Cell count experiments further confirmed the potent inhibitory effects of compounds 3e, 3b, and 3c on MCF-7 and HCT-116 cell growth. Compound 3e demonstrated a reduction of 55-60 % at GI50 and complete inhibition (100 %) at 2x GI50. Compound 3b exhibited 50-55 % reduction (GI50) and 90-95 % inhibition (2x GI50) in HCT-116 cells. Compound 3c displayed 75-80 % inhibition (2x GI50) and 35-40 % inhibition (GI50) in HCT-116 cells. In-depth mechanistic investigations unveiled valuable insights into the mode of action of compound 3e. The cell-cycle assay demonstrated G2/M phase arrest, DNA damage, and caspase-mediated apoptosis in both MCF-7 and HCT-116 cells. Caspase activation indicated a significant increase in apoptosis following exposure to compound 3e. Furthermore, compound 3e induced reactive oxygen species (ROS) production, influencing HCT-116 and MCF-7 cells differently. Elevated ROS production in HCT-116 cells and distinct effects in MCF-7 cells contribute to a deeper understanding of the cytotoxic mechanisms of compound 3e. Overall, these findings highlight the potential of the investigated compounds, particularly compound 3e, as effective inducers of apoptosis in cancer cells. Mechanistic insights into cell cycle arrest, caspase-mediated apoptosis, and ROS modulation provide a comprehensive understanding of their cytotoxic effects. This study offers significant contribution to the development of promising anticancer agents and their therapeutic applications.
Collapse
Affiliation(s)
- Mustapha Musa
- GSK Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham, Triumph Road, Nottingham, NG7 2TU, UK
- Department of Chemistry, Shehu Shagari College of Education, Sokoto, Sokoto State, Nigeria
| | - Muhammadu Bello
- Department of Chemistry, Shehu Shagari College of Education, Sokoto, Sokoto State, Nigeria
| | - Ernest C Agwamba
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Chemistry, Covenant University, Ota, Nigeria
| |
Collapse
|
14
|
Schneider HE, Schmitt LM, Job A, Lankat-Buttgereit B, Gress T, Buchholz M, Gallmeier E. Synthetic lethality between ATR and POLA1 reveals a potential new target for individualized cancer therapy. Neoplasia 2024; 57:101038. [PMID: 39128273 PMCID: PMC11369380 DOI: 10.1016/j.neo.2024.101038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024]
Abstract
The ATR-CHK1 pathway plays a fundamental role in the DNA damage response and is therefore an attractive target in cancer therapy. The antitumorous effect of ATR inhibitors is at least partly caused by synthetic lethality between ATR and various DNA repair genes. In previous studies, we have identified members of the B-family DNA polymerases as potential lethal partner for ATR, i.e. POLD1 and PRIM1. In this study, we validated and characterized the synthetic lethality between ATR and POLA1. First, we applied a model of ATR-deficient DLD-1 human colorectal cancer cells to confirm synthetic lethality by using chemical POLA1 inhibition. Analyzing cell cycle and apoptotic markers via FACS and Western blotting, we were able to show that apoptosis and S phase arrest contributed to the increased sensitivity of ATR-deficient cancer cells towards POLA1 inhibitors. Importantly, siRNA-mediated POLA1 depletion in ATR-deficient cells caused similar effects in regard to impaired cell viability and cumulation of apoptotic markers, thus excluding toxic effects of chemical POLA1 inhibition. Conversely, we demonstrated that siRNA-mediated POLA1 depletion sensitized several cancer cell lines towards chemical inhibition of ATR and its main effector kinase CHK1. In conclusion, the synthetic lethality between ATR/CHK1 and POLA1 might represent a novel and promising approach for individualized cancer therapy: First, alterations of POLA1 could serve as a screening parameter for increased sensitivity towards ATR and CHK1 inhibitors. Second, alterations in the ATR-CHK1 pathway might predict in increased sensitivity towards POLA1 inhibitors.
Collapse
Affiliation(s)
- Hanna Elisabeth Schneider
- Center for Tumor Biology and Immunology, Department of Gastroenterology, Endocrinology and Metabolism, University Hospital of Marburg, Philipps-University Marburg, Marburg, Germany; Department of Medicine A - Hematology, Oncology and Pneumology, University Hospital Münster, Muenster, Germany
| | - Lisa-Maria Schmitt
- Center for Tumor Biology and Immunology, Department of Gastroenterology, Endocrinology and Metabolism, University Hospital of Marburg, Philipps-University Marburg, Marburg, Germany
| | - Albert Job
- Center for Tumor Biology and Immunology, Department of Gastroenterology, Endocrinology and Metabolism, University Hospital of Marburg, Philipps-University Marburg, Marburg, Germany
| | - Brigitte Lankat-Buttgereit
- Center for Tumor Biology and Immunology, Department of Gastroenterology, Endocrinology and Metabolism, University Hospital of Marburg, Philipps-University Marburg, Marburg, Germany
| | - Thomas Gress
- Center for Tumor Biology and Immunology, Department of Gastroenterology, Endocrinology and Metabolism, University Hospital of Marburg, Philipps-University Marburg, Marburg, Germany
| | - Malte Buchholz
- Center for Tumor Biology and Immunology, Department of Gastroenterology, Endocrinology and Metabolism, University Hospital of Marburg, Philipps-University Marburg, Marburg, Germany
| | - Eike Gallmeier
- Center for Tumor Biology and Immunology, Department of Gastroenterology, Endocrinology and Metabolism, University Hospital of Marburg, Philipps-University Marburg, Marburg, Germany; Department of Internal Medicine II - Gastroenterology, Oncology and Metabolism, Hospital Memmingen, Memmingen, Germany.
| |
Collapse
|
15
|
Lee DJ, Cao Y, Juvekar V, Sauraj, Noh CK, Shin SJ, Liu Z, Kim HM. Development of a small molecule-based two-photon photosensitizer for targeting cancer cells. J Mater Chem B 2024. [PMID: 39469993 DOI: 10.1039/d4tb01706d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Photodynamic therapy (PDT) employing two-photon (TP) excitation is increasingly recognized to induce cell damage selectively in targeted areas, underscoring the importance of developing TP photosensitizers (TP-PSs). In this study, we developed BSe-B, a novel PS that combines a selenium containing dye with biotin, a cancer-selective ligand, and is optimized for TP excitation. BSe-B demonstrated enhanced cancer selectivity, efficient generation of type-I based reactive oxygen species (ROS), low dark toxicity, and excellent cell-staining capability. Evaluation across diverse cell lines (HeLa, A549, OVCAR-3, WI-38, and L-929) demonstrated that BSe-B differentiated and targeted cancer cells while sparing normal cells. BSe-B displayed excellent in vivo biocompatibility. In cancer models such as three-dimensional spheroids and actual colon cancer tissues, BSe-B selectively induced ROS production and cell death under TP irradiation, demonstrating precise spatial control. These findings highlight the potential of BSe-B for imaging-guided PDT and its capability for micro treatment within tissues. Thus, BSe-B demonstrates robust TP-PDT capabilities, making it a promising dual-purpose tool for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Dong Joon Lee
- Department of Energy Systems Research and Department of Chemistry, Ajou University, Suwon 16499, Korea.
| | - Yu Cao
- College of Health Science and Engineering, Hubei University, Wuhan 430062, China.
| | - Vinayak Juvekar
- Department of Energy Systems Research and Department of Chemistry, Ajou University, Suwon 16499, Korea.
| | - Sauraj
- Department of Energy Systems Research and Department of Chemistry, Ajou University, Suwon 16499, Korea.
| | - Choong-Kyun Noh
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Sung Jae Shin
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Zhihong Liu
- College of Health Science and Engineering, Hubei University, Wuhan 430062, China.
| | - Hwan Myung Kim
- Department of Energy Systems Research and Department of Chemistry, Ajou University, Suwon 16499, Korea.
| |
Collapse
|
16
|
McRobb LS, Lee VS, Faqihi F, Stoodley MA. A Simple Model to Study Mosaic Gene Expression in 3D Endothelial Spheroids. J Cardiovasc Dev Dis 2024; 11:305. [PMID: 39452276 PMCID: PMC11508842 DOI: 10.3390/jcdd11100305] [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: 06/07/2024] [Revised: 09/13/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
AIMS The goal of this study was to establish a simple model of 3D endothelial spheroids with mosaic gene expression using adeno-associated virus (AAV) transduction, with a future aim being to study the activity of post-zygotic mutations common to vascular malformations. METHODS In this study, 96-well U-bottom plates coated with a commercial repellent were seeded with two immortalized human endothelial cell lines and aggregation monitored using standard microscopy or live-cell analysis. The eGFP expression was used to monitor the AAV transduction. RESULTS HUVEC-TERT2 could not form spheroids spontaneously. The inclusion of collagen I in the growth medium could stimulate cell aggregation; however, these spheroids were not stable. In contrast, the hCMEC/D3 cells aggregated spontaneously and formed reproducible, robust 3D spheroids within 3 days, growing steadily for at least 4 weeks without the need for media refreshment. The hCMEC/D3 spheroids spontaneously developed a basement membrane, including collagen I, and expressed endothelial-specific CD31 at the spheroid surface. Serotypes AAV1 and AAV2QUADYF transduced these spheroids without toxicity and established sustained, mosaic eGFP expression. CONCLUSIONS In the future, this simple approach to endothelial spheroid formation combined with live-cell imaging could be used to rapidly assess the 3D phenotypes and drug and radiation sensitivities arising from mosaic mutations common to brain vascular malformations.
Collapse
Affiliation(s)
- Lucinda S. McRobb
- Macquarie Medical School, Faculty of Medicine, Health, and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia (M.A.S.)
| | | | | | | |
Collapse
|
17
|
Dias IRSB, Costa RGA, Rodrigues ACBDC, Silva SLR, Oliveira MDS, Soares MBP, Dias RB, Valverde LF, Gurgel Rocha CA, Cairns LV, Mills KI, Bezerra DP. Bithionol eliminates acute myeloid leukaemia stem-like cells by suppressing NF-κB signalling and inducing oxidative stress, leading to apoptosis and ferroptosis. Cell Death Discov 2024; 10:390. [PMID: 39209810 PMCID: PMC11362533 DOI: 10.1038/s41420-024-02148-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Acute myeloid leukaemia (AML) is a lethal bone marrow neoplasm caused by genetic alterations in blood cell progenitors. Leukaemic stem cells (LSCs) are responsible for the development of AML, drug resistance and relapse. Bithionol is an old anthelmintic drug with potential antibacterial, antiviral, antifungal, anti-Alzheimer, and antitumour properties. In this work, we focused on the anti-AML LSC properties of bithionol. This compound inhibited the viability of both solid and haematological cancer cells, suppressed AML stem-like cells, and inhibited AML growth in NSG mice at a dosage of 50 mg/kg, with tolerable systemic toxicity. Bithionol significantly reduced the levels of phospho-NF-κB p65 (Ser529) and phospho-NF-κB p65 (Ser536) and nuclear NF-κB p65 translocation in AML cells, indicating that this molecule can suppress NF-κB signalling. DNA fragmentation, nuclear condensation, cell shrinkage, phosphatidylserine externalisation, loss of transmembrane mitochondrial potential, caspase-3 activation and PARP-(Asp 214) cleavage were detected in bithionol-treated AML cells, indicating the induction of apoptosis. Furthermore, this compound increased mitochondrial superoxide levels, and bithionol-induced cell death was partially prevented by cotreatment with the selective ferroptosis inhibitor ferrostatin-1, indicating the induction of ferroptosis. In addition, bithionol synergised with venetoclax in AML cells, indicating the translational potential of bithionol to enhance the effects of venetoclax in patients with AML. Taken together, these data indicate that bithionol is a potential new anti-AML drug.
Collapse
Affiliation(s)
- Ingrid R S B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, Brazil
| | - Rafaela G A Costa
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, Brazil
| | | | - Suellen L R Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, Brazil
| | - Maiara de S Oliveira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, Brazil
- SENAI Institute for Innovation in Advanced Health Systems, SENAI CIMATEC, Salvador, BA, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, Brazil
- Department of Propaedeutics, Faculty of Dentistry of the Federal University of Bahia (UFBA), Salvador, Bahia, Brazil
- Department of Biological Sciences, State University of Feira de Santana, Feira de Santana, Bahia, Brazil
| | - Ludmila F Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, Brazil
- Department of Dentistry, Federal University of Sergipe, Lagarto, Sergipe, Brazil
| | - Clarissa A Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, Brazil
- Department of Propaedeutics, Faculty of Dentistry of the Federal University of Bahia (UFBA), Salvador, Bahia, Brazil
- Center for Biotechnology and Cell Therapy, D'Or Institute for Research and Education (IDOR), Salvador, Bahia, Brazil
| | - Lauren V Cairns
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Ken I Mills
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, Brazil.
| |
Collapse
|
18
|
Gonçalves GR, Teixeira T, Bezerra DP, Soares MBP, Silva VR, Santos LDS, Batista AA, Oliveira KM, Correa RS. Exploring the BSA- and DNA-binding, cytotoxicity, and cell cycle evaluation of ternary copper(II)/diimine complexes with N, N-dibenzyl- N'-benzoylthiourea as promising metallodrug candidates. Dalton Trans 2024; 53:12951-12961. [PMID: 38842058 DOI: 10.1039/d4dt01152j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Four new copper(II) complexes were synthesized and characterized with the general formula [Cu(N-N)(Th)(NO3)], where N-N corresponds to the N-heterocyclic ligands 1,10-phenanthroline (phen), 2,2'-bipyridine (bipy), 4,7-diphenyl-1,10-phenanthroline (dpp), and 4,4-dimethyl-2,2'-bipyridine (dmbp) and Th represents the N,N-dibenzyl-N'-benzoylthiourea. Cytotoxic activities of the complexes against HCT116 (human colon carcinoma), HepG2 (human hepatocellular carcinoma), and non-tumor MRC-5 (human lung fibroblast) cells were investigated. The copper(II) complexes 1-4 were characterized by spectroscopic techniques while complexes 1 and 2 were studied using single-crystal X-ray diffraction as well. The complexes possessed a five-coordinated structure with one nitrate ligand as a monodentate at the axial position and two bidentate ligands N-heterocyclic and N,N-dibenzyl-N'-benzoylthiourea. The complexes showed promising IC50 values, ranging from 0.3 to 9.0 μM. Furthermore, interaction studies with biomolecules such as calf thymus DNA (ct-DNA) and Bovine Serum Albumin (BSA), which can act as possible biological targets of the complexes, were carried out. The studies suggested that the compounds interact moderately with ct-DNA and BSA. Complexes 1, 2, and 4 did not lead to cell accumulation at any stage of the cell cycle but caused a significant increase in internucleosomal DNA fragmentation. Whereas, compound 3 caused cell cycle arrest in the S phase while doxorubicin caused cell cycle arrest in the G2/M phase. The effect of structural modifications on the metal compounds was correlated with their biological properties and it was concluded that an increase in biological activity occurred with increasing the extension of the diimine ligands. Thus, complex 3 was the most promising one.
Collapse
Affiliation(s)
- Guilherme R Gonçalves
- Departamento de Química, ICEB, Universidade Federal de Ouro Preto - UFOP, CEP 35400-000, Ouro Preto, MG, Brazil.
| | - Tamara Teixeira
- Departamento de Química, ICEB, Universidade Federal de Ouro Preto - UFOP, CEP 35400-000, Ouro Preto, MG, Brazil.
| | - Daniel P Bezerra
- Instituto Gonçalo Moniz - Fundação Oswaldo Cruz (IGM-FIOCRUZ-BA), CEP 40296-710, Salvador, BA, Brazil
| | - Milena B P Soares
- Instituto Gonçalo Moniz - Fundação Oswaldo Cruz (IGM-FIOCRUZ-BA), CEP 40296-710, Salvador, BA, Brazil
| | - Valdenizia R Silva
- Instituto Gonçalo Moniz - Fundação Oswaldo Cruz (IGM-FIOCRUZ-BA), CEP 40296-710, Salvador, BA, Brazil
| | - Luciano de S Santos
- Instituto Gonçalo Moniz - Fundação Oswaldo Cruz (IGM-FIOCRUZ-BA), CEP 40296-710, Salvador, BA, Brazil
| | - Alzir A Batista
- Departamento de Química, Universidade Federal de São Carlos - UFSCar, CP 676, CEP 13561-901, São Carlos, SP, Brazil
| | - Katia M Oliveira
- Departamento de Química, ICEB, Universidade Federal de Ouro Preto - UFOP, CEP 35400-000, Ouro Preto, MG, Brazil.
- Instituto de Química, Universidade de Brasília - UnB, Campus Darcy Ribeiro, CEP 70910-900, Brasília, DF, Brazil
| | - Rodrigo S Correa
- Departamento de Química, ICEB, Universidade Federal de Ouro Preto - UFOP, CEP 35400-000, Ouro Preto, MG, Brazil.
| |
Collapse
|
19
|
Neves SP, Bomfim LM, Kataura T, Carvalho SG, Nogueira ML, Dias RB, Valverde LDF, Gurgel Rocha CA, Soares MBP, Silva MMD, Batista AA, Korolchuk VI, Bezerra DP. Ruthenium complex containing 1,3-thiazolidine-2-thione inhibits hepatic cancer stem cells by suppressing Akt/mTOR signalling and leading to apoptotic and autophagic cell death. Biomed Pharmacother 2024; 177:117059. [PMID: 38955086 DOI: 10.1016/j.biopha.2024.117059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/16/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
Hepatic cancer is one of the main causes of cancer-related death worldwide. Cancer stem cells (CSCs) are a unique subset of cancer cells that promote tumour growth, maintenance, and therapeutic resistance, leading to recurrence. In the present work, the ability of a ruthenium complex containing 1,3-thiazolidine-2-thione (RCT), with the chemical formula [Ru(tzdt)(bipy)(dppb)]PF6, to inhibit hepatic CSCs was explored in human hepatocellular carcinoma HepG2 cells. RCT exhibited potent cytotoxicity to solid and haematological cancer cell lines and reduced the clonogenic potential, CD133+ and CD44high cell percentages and tumour spheroid growth of HepG2 cells. RCT also inhibited cell motility, as observed in the wound healing assay and transwell cell migration assay. RCT reduced the levels of Akt1, phospho-Akt (Ser473), phospho-Akt (Thr308), phospho-mTOR (Ser2448), and phospho-S6 (Ser235/Ser236) in HepG2 cells, indicating that interfering with Akt/mTOR signalling is a mechanism of action of RCT. The levels of active caspase-3 and cleaved PARP (Asp214) were increased in RCT-treated HepG2 cells, indicating the induction of apoptotic cell death. In addition, RCT modulated the autophagy markers LC3B and p62/SQSTM1 in HepG2 cells and increased mitophagy in a mt-Keima-transfected mouse embryonic fibroblast (MEF) cell model, and RCT-induced cytotoxicity was partially prevented by autophagy inhibitors. Furthermore, mutant Atg5-/- MEFs and PentaKO HeLa cells (human cervical adenocarcinoma with five autophagy receptor knockouts) were less sensitive to RCT cytotoxicity than their parental cell lines, indicating that RCT induces autophagy-mediated cell death. Taken together, these data indicate that RCT is a novel potential anti-liver cancer drug with a suppressive effect on CSCs.
Collapse
Affiliation(s)
- Sara P Neves
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Larissa M Bomfim
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Tetsushi Kataura
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Sabrine G Carvalho
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Mateus L Nogueira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil; Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador, Bahia, 40110-909, Brazil; Department of Biological Sciences, State University of Feira de Santana, Feira de Santana, Bahia, 44036-900, Brazil
| | - Ludmila de F Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil; Department of Dentistry, Federal University of Sergipe, Lagarto, Sergipe, 49400-000, Brazil
| | - Clarissa A Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil; Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador, Bahia, 40110-909, Brazil; Center for Biotechnology and Cell Therapy, D'Or Institute for Research and Education (IDOR), Salvador, Bahia, 41253-190, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil; SENAI Institute of Innovation (ISI) in Health Advanced Systems, University Center SENAI/CIMATEC, Salvador, Bahia, 41650-010, Brazil
| | - Monize M da Silva
- Department of Chemistry, Federal University of São Carlos, São Carlos, São Paulo, 13561-901 Brazil
| | - Alzir A Batista
- Department of Chemistry, Federal University of São Carlos, São Carlos, São Paulo, 13561-901 Brazil
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil.
| |
Collapse
|
20
|
Burgers LD, Ciurus S, Engel P, Kuntschar S, Raue R, Kiprina A, Primke T, Schmid T, Weigert A, Schmidtko A, Fürst R. (Homo-)harringtonine prevents endothelial inflammation through IRF-1 dependent downregulation of VCAM1 mRNA expression and inhibition of cell adhesion molecule protein biosynthesis. Biomed Pharmacother 2024; 176:116907. [PMID: 38865849 DOI: 10.1016/j.biopha.2024.116907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024] Open
Abstract
The plant alkaloid homoharringtonine (HHT) is a Food and Drug Administration (FDA)-approved drug for the treatment of hematologic malignancies. In addition to its well-established antitumor activity, accumulating evidence attributes anti-inflammatory effects to HHT, which have mainly been studied in leukocytes to date. However, a potential influence of HHT on inflammatory activation processes in endothelial cells, which are a key feature of inflammation and a prerequisite for the leukocyte-endothelial cell interaction and leukocyte extravasation, remains poorly understood. In this study, the anti-inflammatory potential of HHT and its derivative harringtonine (HT) on the TNF-induced leukocyte-endothelial cell interaction was assessed, and the underlying mechanistic basis of these effects was elucidated. HHT affected inflammation in vivo in a murine peritonitis model by reducing leukocyte infiltration and proinflammatory cytokine expression as well as ameliorating abdominal pain behavior. In vitro, HT and HHT impaired the leukocyte-endothelial cell interaction by decreasing the expression of the endothelial cell adhesion molecules intracellular adhesion molecule -1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1). This effect was mediated by a bipartite mechanism. While HHT did not affect the prominent TNF-induced pro-inflammatory NF-ĸB signaling cascade, the compound downregulated the VCAM1 mRNA expression in an IRF-1-dependent manner and diminished active ICAM1 mRNA translation as determined by polysome profiling. This study highlights HHT as an anti-inflammatory compound that efficiently hampers the leukocyte-endothelial cell interaction by targeting endothelial activation processes.
Collapse
Affiliation(s)
- Luisa D Burgers
- Institute of Pharmaceutical Biology, Faculty of Biochemistry, Chemistry and Pharmacy, Goethe University, Frankfurt am Main, Germany
| | - Sarah Ciurus
- Institute of Pharmaceutical Biology, Faculty of Biochemistry, Chemistry and Pharmacy, Goethe University, Frankfurt am Main, Germany
| | - Patrick Engel
- Institute of Pharmacology and Clinical Pharmacy, Faculty of Biochemistry, Chemistry and Pharmacy, Goethe University, Frankfurt am Main, Germany
| | - Silvia Kuntschar
- Institute of Biochemistry I, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Rebecca Raue
- Institute of Biochemistry I, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Anastasiia Kiprina
- Institute of Biochemistry I, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Tobias Primke
- Institute of Pharmaceutical Biology, Faculty of Biochemistry, Chemistry and Pharmacy, Goethe University, Frankfurt am Main, Germany
| | - Tobias Schmid
- Institute of Biochemistry I, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Achim Schmidtko
- Institute of Pharmacology and Clinical Pharmacy, Faculty of Biochemistry, Chemistry and Pharmacy, Goethe University, Frankfurt am Main, Germany
| | - Robert Fürst
- Institute of Pharmaceutical Biology, Faculty of Biochemistry, Chemistry and Pharmacy, Goethe University, Frankfurt am Main, Germany; LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), Frankfurt am Main, Germany; Pharmaceutical Biology, Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
21
|
Solyanik G, Kolesnik D, Prokhorova I, Yurchenko O, Pyaskovskaya O. Mitochondrial dysfunction significantly contributes to the sensitivity of tumor cells to anoikis and their metastatic potential. Heliyon 2024; 10:e32626. [PMID: 38994085 PMCID: PMC11237942 DOI: 10.1016/j.heliyon.2024.e32626] [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: 11/19/2023] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 07/13/2024] Open
Abstract
It is well-known that the survival of metastatic cells during their dissemination plays an important role in metastasis. However, does this mean that the final result of the metastatic cascade (the volume of metastatic damage to distant organs and tissues) depends on, or at least correlates with, the degree of resistance to anoikis (distinctive hallmarks of metastatic cells)? This question remains open.The aim of the work was to study in vitro the changes in the survival rates, proliferative activity, oxidative stress, and glycolysis intensity during three days of anchorage-dependent and anchorage-independent growth of two Lewis lung carcinoma cell lines (LLC and LLC/R9) and compare these changes with the status of mitochondria and metastatic potential of the cells in vivo. Methods The number and volume of lung metastases were estimated for each cell line after intramuscular inoculation of the cells in C57Bl/6 mice. For the in vitro study, the cells were seeded on Petri dishes pretreated with poly-HEMA or untreated dishes and then allowed to grow for 3 days. Cell viability, cell cycle progression, the level of reactive oxygen species (ROS), glucose consumption and lactate production rates were investigated daily in both growth conditions. An electron microscopy study of intracellular structures was carried out. Results The study showed (as far as we know for the first time) a correlation between the metastatic potential of cells (determined in vivo) and their sensitivity to anoikis (assessed in vitro). The transition of LLC/R9 cells with an inherently defective mitochondrial system to the conditions of anchorage-independent growth was characterized by a decrease in survival, a slowdown in growth rates, an increase in both glucose consumption rate and intracellular ROS levels and manyfold lower metastatic potential, compared to highly metastatic LLC cells with the normal mitochondrial system.
Collapse
Affiliation(s)
- G.I. Solyanik
- Laboratory of Molecular and Cellular Mechanisms of Metastasis, RE Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, The National Academy of Sciences of Ukraine, Kyiv, 03022, Ukraine
| | - D.L. Kolesnik
- Laboratory of Molecular and Cellular Mechanisms of Metastasis, RE Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, The National Academy of Sciences of Ukraine, Kyiv, 03022, Ukraine
| | - I.V. Prokhorova
- Laboratory of Molecular and Cellular Mechanisms of Metastasis, RE Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, The National Academy of Sciences of Ukraine, Kyiv, 03022, Ukraine
| | - O.V. Yurchenko
- Laboratory of Molecular and Cellular Mechanisms of Metastasis, RE Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, The National Academy of Sciences of Ukraine, Kyiv, 03022, Ukraine
| | - O.N. Pyaskovskaya
- Laboratory of Molecular and Cellular Mechanisms of Metastasis, RE Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, The National Academy of Sciences of Ukraine, Kyiv, 03022, Ukraine
| |
Collapse
|
22
|
Buratta S, Urbanelli L, Pellegrino RM, Alabed HBR, Latella R, Cerrotti G, Emiliani C, Bassotti G, Spaterna A, Marconi P, Fettucciari K. PhosphoLipidome Alteration Induced by Clostridioides difficile Toxin B in Enteric Glial Cells. Cells 2024; 13:1103. [PMID: 38994956 PMCID: PMC11240607 DOI: 10.3390/cells13131103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
Clostridioides difficile (C. difficile) is responsible for a spectrum of nosocomial/antibiotic-associated gastrointestinal diseases that are increasing in global incidence and mortality rates. The C. difficile pathogenesis is due to toxin A and B (TcdA/TcdB), both causing cytopathic and cytotoxic effects and inflammation. Recently, we demonstrated that TcdB induces cytopathic and cytotoxic (apoptosis and necrosis) effects in enteric glial cells (EGCs) in a dose/time-dependent manner and described the underlying signaling. Despite the role played by lipids in host processes activated by pathogens, to counter infection and/or induce cell death, to date no studies have investigated lipid changes induced by TcdB/TcdA. Here, we evaluated the modification of lipid composition in our in vitro model of TcdB infection. Apoptosis, cell cycle, cell viability, and lipidomic profiles were evaluated in EGCs treated for 24 h with two concentrations of TcdB (0.1 ng/mL; 10 ng/mL). In EGCs treated with the highest concentration of TcdB, not only an increased content of total lipids was observed, but also lipidome changes, allowing the separation of TcdB-treated cells and controls into different clusters. The statistical analyses also allowed us to ascertain which lipid classes and lipid molecular species determine the clusterization. Changes in lipid species containing inositol as polar head and plasmalogen phosphatidylethanolamine emerged as key indicators of altered lipid metabolism in TcdB-treated EGCs. These results not only provide a picture of the phospholipid profile changes but also give information regarding the lipid metabolism pathways altered by TcdB, and this might represent an important step for developing strategies against C. difficile infection.
Collapse
Affiliation(s)
- Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (S.B.); (L.U.); (R.M.P.); (H.B.R.A.); (R.L.); (G.C.); (C.E.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (S.B.); (L.U.); (R.M.P.); (H.B.R.A.); (R.L.); (G.C.); (C.E.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Roberto Maria Pellegrino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (S.B.); (L.U.); (R.M.P.); (H.B.R.A.); (R.L.); (G.C.); (C.E.)
| | - Husam B. R. Alabed
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (S.B.); (L.U.); (R.M.P.); (H.B.R.A.); (R.L.); (G.C.); (C.E.)
| | - Raffaella Latella
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (S.B.); (L.U.); (R.M.P.); (H.B.R.A.); (R.L.); (G.C.); (C.E.)
| | - Giada Cerrotti
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (S.B.); (L.U.); (R.M.P.); (H.B.R.A.); (R.L.); (G.C.); (C.E.)
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (S.B.); (L.U.); (R.M.P.); (H.B.R.A.); (R.L.); (G.C.); (C.E.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Gabrio Bassotti
- Department of Medicine and Surgery, Gastroenterology, Hepatology & Digestive Endoscopy Section, University of Perugia, Piazzale Lucio Severi 1, 06132 Perugia, Italy;
- Santa Maria Della Misericordia Hospital, Gastroenterology & Hepatology Unit, Piazzale Menghini 1, 06129 Perugia, Italy
| | - Andrea Spaterna
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Macerata, Italy
| | - Pierfrancesco Marconi
- Department of Medicine and Surgery, Biosciences & Medical Embryology Section, University of Perugia, Piazzale Lucio Severi 1, 06132 Perugia, Italy;
| | - Katia Fettucciari
- Department of Medicine and Surgery, Biosciences & Medical Embryology Section, University of Perugia, Piazzale Lucio Severi 1, 06132 Perugia, Italy;
| |
Collapse
|
23
|
Schmitt L, Hoppe J, Cea-Medina P, Bruch PM, Krings KS, Lechtenberg I, Drießen D, Peter C, Bhatia S, Dietrich S, Stork B, Fritz G, Gohlke H, Müller TJJ, Wesselborg S. Novel meriolin derivatives potently inhibit cell cycle progression and transcription in leukemia and lymphoma cells via inhibition of cyclin-dependent kinases (CDKs). Cell Death Discov 2024; 10:279. [PMID: 38862521 PMCID: PMC11167047 DOI: 10.1038/s41420-024-02056-6] [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: 04/24/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024] Open
Abstract
A key feature of cancer is the disruption of cell cycle regulation, which is characterized by the selective and abnormal activation of cyclin-dependent kinases (CDKs). Consequently, targeting CDKs via meriolins represents an attractive therapeutic approach for cancer therapy. Meriolins represent a semisynthetic compound class derived from meridianins and variolins with a known CDK inhibitory potential. Here, we analyzed the two novel derivatives meriolin 16 and meriolin 36 in comparison to other potent CDK inhibitors and could show that they displayed a high cytotoxic potential in different lymphoma and leukemia cell lines as well as in primary patient-derived lymphoma and leukemia cells. In a kinome screen, we showed that meriolin 16 and 36 prevalently inhibited most of the CDKs (such as CDK1, 2, 3, 5, 7, 8, 9, 12, 13, 16, 17, 18, 19, 20). In drug-to-target modeling studies, we predicted a common binding mode of meriolin 16 and 36 to the ATP-pocket of CDK2 and an additional flipped binding for meriolin 36. We could show that cell cycle progression and proliferation were blocked by abolishing phosphorylation of retinoblastoma protein (a major target of CDK2) at Ser612 and Thr82. Moreover, meriolin 16 prevented the CDK9-mediated phosphorylation of RNA polymerase II at Ser2 which is crucial for transcription initiation. This renders both meriolin derivatives as valuable anticancer drugs as they target three different Achilles' heels of the tumor: (1) inhibition of cell cycle progression and proliferation, (2) prevention of transcription, and (3) induction of cell death.
Collapse
Affiliation(s)
- Laura Schmitt
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Julia Hoppe
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Pablo Cea-Medina
- Institute for Pharmaceutical and Medicinal Chemistry, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Peter-Martin Bruch
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany
- Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Karina S Krings
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Ilka Lechtenberg
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Daniel Drießen
- Institute of Organic Chemistry and Macromolecular Chemistry, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Christoph Peter
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Sanil Bhatia
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Sascha Dietrich
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany
- Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Björn Stork
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Gerhard Fritz
- Institute of Toxicology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Holger Gohlke
- Institute for Pharmaceutical and Medicinal Chemistry, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
- John von Neumann Institute for Computing (NIC), Jülich Supercomputing Center (JSC) and Institute of Bio- and Geosciences (IBG-4: Bioinformatics), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52425, Jülich, Germany
| | - Thomas J J Müller
- Institute of Organic Chemistry and Macromolecular Chemistry, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Sebastian Wesselborg
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany.
- Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf (CIO ABCD), Düsseldorf, Germany.
| |
Collapse
|
24
|
Al-Hayali MZ, Nge CE, Lim KH, Collins HM, Kam TS, Bradshaw TD. Conofolidine: A Natural Plant Alkaloid That Causes Apoptosis and Senescence in Cancer Cells. Molecules 2024; 29:2654. [PMID: 38893527 PMCID: PMC11173856 DOI: 10.3390/molecules29112654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Natural products contribute substantially to anticancer therapy; the plant kingdom provides an important source of molecules. Conofolidine is a novel Aspidosperma-Aspidosperma bisindole alkaloid isolated from the Malayan plant Tabernaemontana corymbosa. Herein, we report conofolidine's broad-spectrum anticancer activity together with that of three other bisindoles-conophylline, leucophyllidine, and bipleiophylline-against human-derived breast, colorectal, pancreatic, and lung carcinoma cell lines. Remarkably, conofolidine was able to induce apoptosis (e.g., in MDA-MB-468 breast) or senescence (e.g., in HT-29 colorectal) in cancer cells. Annexin V-FITC/PI, caspase activation, and PARP cleavage confirmed the former while positive β-gal staining corroborated the latter. Cell cycle perturbations were evident, comprising S-phase depletion, accompanied by downregulated CDK2, and cyclins (A2, D1) with p21 upregulation. Confocal imaging of HCT-116 cells revealed an induction of aberrant mitotic phenotypes-membrane blebbing, DNA-fragmentation with occasional multi-nucleation. DNA integrity assessment in HCT-116, MDA-MB-468, MIAPaCa-2, and HT-29 cells showed increased fluorescent γ-H2AX during the G1 cell cycle phase; γ-H2AX foci were validated in HCT-116 and MDA-MB-468 cells by confocal microscopy. Conofolidine increased oxidative stress, preceding apoptosis- and senescence-induction in most carcinoma cell lines as seen by enhanced ROS levels accompanied by increased NQO1 expression. Collectively, we present conofolidine as a putative potent anticancer agent capable of inducing heterogeneous modes of cancerous cell death in vitro, encouraging further preclinical evaluations of this natural product.
Collapse
Affiliation(s)
- Mohammed Zuhair Al-Hayali
- School of Pharmacy, Al-Kitab University, Kirkuk 36015, Iraq
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, University Park, Nottingham NG7 2RD, UK;
| | - Choy-Eng Nge
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia; (C.-E.N.); (T.-S.K.)
| | - Kuan Hon Lim
- School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, Semenyih 43500, Malaysia;
| | - Hilary M. Collins
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, University Park, Nottingham NG7 2RD, UK;
| | - Toh-Seok Kam
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia; (C.-E.N.); (T.-S.K.)
| | - Tracey D. Bradshaw
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, University Park, Nottingham NG7 2RD, UK;
| |
Collapse
|
25
|
Bomfim LM, Neves SP, Coelho AMRM, Nogueira ML, Dias RB, Valverde LDF, Rocha CAG, Soares MBP, Batista AA, Correa RS, Bezerra DP. Ru(II)-based complexes containing 2-thiouracil derivatives suppress liver cancer stem cells by targeting NF-κB and Akt/mTOR signaling. Cell Death Discov 2024; 10:270. [PMID: 38830859 PMCID: PMC11148080 DOI: 10.1038/s41420-024-02036-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
Cancer stem cells (CSCs) are defined as a rare population of cancer cells related to tumor initiation and maintenance. These cells are primarily responsible for tumor growth, invasion, metastasis, recurrence, and resistance to chemotherapy. In this paper, we demonstrated the ability of Ru(II)-based complexes containing 2-thiouracil derivatives with the chemical formulas trans-[Ru(2TU)(PPh3)2(bipy)]PF6 (1) and trans-[Ru(6m2TU)(PPh3)2(bipy)]PF6 (2) (where 2TU = 2-thiouracil and 6m2TU = 6-methyl-2-thiouracil) to suppress liver CSCs by targeting NF-κB and Akt/mTOR signaling. Complexes 1 and 2 displayed potent cytotoxic effects on cancer cell lines and suppressed liver CSCs from HepG2 cells. Increased phosphatidylserine exposure, loss of mitochondrial transmembrane potential, increased PARP (Asp214) cleavage, DNA fragmentation, chromatin condensation and cytoplasmic shrinkage were detected in HepG2 cells treated with these complexes. Mechanistically, complexes 1 and 2 target NF-κB and Akt/mTOR signaling in HepG2 cells. Cell motility inhibition was also detected in HepG2 cells treated with these complexes. Complexes 1 and 2 also inhibited tumor progression in mice with HepG2 cell xenografts and exhibited tolerable systemic toxicity. Taken together, these results indicate that these complexes are new anti-HCC drug candidates that can suppress liver CSCs.
Collapse
Affiliation(s)
- Larissa M Bomfim
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Sara P Neves
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Amanda M R M Coelho
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Mateus L Nogueira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador, Bahia, 40110-909, Brazil
- Department of Biological Sciences, State University of Feira de Santana, Feira de Santana, Bahia, 44036-900, Brazil
| | - Ludmila de F Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
- Department of Dentistry, Federal University of Sergipe, Lagarto, Sergipe, 49400-000, Brazil
| | - Clarissa A G Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador, Bahia, 40110-909, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador, Bahia, 40110-909, Brazil
- Center for Biotechnology and Cell Therapy, D'Or Institute for Research and Education (IDOR), Salvador, Bahia, 41253-190, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
- SENAI Institute of Innovation (ISI) in Health Advanced Systems, University Center SENAI/CIMATEC, Salvador, Bahia, 41650-010, Brazil
| | - Alzir A Batista
- Department of Chemistry, Federal University of São Carlos, São Paulo, São Carlos, 13561-901, Brazil
| | - Rodrigo S Correa
- Department of Chemistry, Federal University of Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil.
| |
Collapse
|
26
|
Zech TJ, Wolf A, Hector M, Bischoff-Kont I, Krishnathas GM, Kuntschar S, Schmid T, Bracher F, Langmann T, Fürst R. 2-Desaza-annomontine (C81) impedes angiogenesis through reduced VEGFR2 expression derived from inhibition of CDC2-like kinases. Angiogenesis 2024; 27:245-272. [PMID: 38403816 PMCID: PMC11021337 DOI: 10.1007/s10456-024-09906-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/15/2024] [Indexed: 02/27/2024]
Abstract
Angiogenesis is a crucial process in the progression of various pathologies, like solid tumors, wet age-related macular degeneration, and chronic inflammation. Current anti-angiogenic treatments still have major drawbacks like limited efficacy in diseases that also rely on inflammation. Therefore, new anti-angiogenic approaches are sorely needed, and simultaneous inhibition of angiogenesis and inflammation is desirable. Here, we show that 2-desaza-annomontine (C81), a derivative of the plant alkaloid annomontine previously shown to inhibit endothelial inflammation, impedes angiogenesis by inhibiting CDC2-like kinases (CLKs) and WNT/β-catenin signaling. C81 reduced choroidal neovascularization in a laser-induced murine in vivo model, inhibited sprouting from vascular endothelial growth factor A (VEGF-A)-activated murine aortic rings ex vivo, and reduced angiogenesis-related activities of endothelial cells in multiple functional assays. This was largely phenocopied by CLK inhibitors and knockdowns, but not by inhibitors of the other known targets of C81. Mechanistically, CLK inhibition reduced VEGF receptor 2 (VEGFR2) mRNA and protein expression as well as downstream signaling. This was partly caused by a reduction of WNT/β-catenin pathway activity, as activating the pathway induced, while β-catenin knockdown impeded VEGFR2 expression. Surprisingly, alternative splicing of VEGFR2 was not detected. In summary, C81 and other CLK inhibitors could be promising compounds in the treatment of diseases that depend on angiogenesis and inflammation due to their impairment of both processes.
Collapse
Affiliation(s)
- T J Zech
- Faculty of Biochemistry, Chemistry and Pharmacy, Institute for Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany.
| | - A Wolf
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - M Hector
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - I Bischoff-Kont
- Faculty of Biochemistry, Chemistry and Pharmacy, Institute for Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - G M Krishnathas
- Faculty of Biochemistry, Chemistry and Pharmacy, Institute for Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - S Kuntschar
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - T Schmid
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - F Bracher
- Pharmaceutical Chemistry, Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - T Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - R Fürst
- Faculty of Biochemistry, Chemistry and Pharmacy, Institute for Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
- Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
27
|
Bennett HA, McAdorey A, Yan H. Staining Properties of Selected Commercial Fluorescent Dyes Toward B- and Z-DNA. J Fluoresc 2024; 34:1193-1205. [PMID: 37505363 DOI: 10.1007/s10895-023-03343-8] [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: 06/03/2023] [Accepted: 07/06/2023] [Indexed: 07/29/2023]
Abstract
The properties of six commonly used, commercially available, fluorescent dyes were compared in staining right-handed B-DNA and left-handed Z-DNA. All showed different degree of fluorescence turn-on in the presence of B-DNA, but very little in the presence of Z-DNA. The optimal range of dye-DNA ratios of DNA was determined. While these dyes do not provide a turn-on type probe for Z-DNA, staining between B- and Z-DNA using dyes such as SYBR Green I was shown to be useful in tracking the kinetics of conformational changes between these two forms of DNA. Finally, SYBR Green I showed unique circular dichroism patterns in 4 M NaCl that change in the presence of double stranded DNA, both in the visible and UV range.
Collapse
Affiliation(s)
- Hayley-Ann Bennett
- Department of Chemistry and Centre for Biotechnology, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, L2S 3A1, Canada
| | - Alyssa McAdorey
- Department of Chemistry and Centre for Biotechnology, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, L2S 3A1, Canada
| | - Hongbin Yan
- Department of Chemistry and Centre for Biotechnology, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, L2S 3A1, Canada.
| |
Collapse
|
28
|
Silva SLR, Dias IRSB, Rodrigues ACBDC, Costa RGA, Oliveira MDS, Barbosa GADC, Soares MBP, Dias RB, Valverde LF, Rocha CAG, Roy N, Park CY, Bezerra DP. Emetine induces oxidative stress, cell differentiation and NF-κB inhibition, suppressing AML stem/progenitor cells. Cell Death Discov 2024; 10:201. [PMID: 38684672 PMCID: PMC11059384 DOI: 10.1038/s41420-024-01967-8] [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: 02/21/2024] [Revised: 04/09/2024] [Accepted: 04/15/2024] [Indexed: 05/02/2024] Open
Abstract
Acute myeloid leukemia (AML) is a fatal malignancy of the blood and bone marrow. Leukemic stem cells (LSCs) are a rare subset of leukemic cells that promote the development and progression of AML, and eradication of LSCs is critical for effective control of this disease. Emetine is an FDA-approved antiparasitic drug with antitumor properties; however, little is known about its potential against LSCs. Herein, we explored the antileukemic potential of emetine, focusing on its effects on AML stem/progenitor cells. Emetine exhibited potent cytotoxic activity both in hematologic and solid cancer cells and induced AML cell differentiation. Emetine also inhibited AML stem/progenitor cells, as evidenced by decreased expression of CD34, CD97, CD99, and CD123 in KG-1a cells, indicating anti-AML stem/progenitor cell activities. The administration of emetine at a dosage of 10 mg/kg for two weeks showed no significant toxicity and significantly reduced xenograft leukemic growth in vivo. NF-κB activation was reduced in emetine-treated KG-1a cells, as shown by reduced phospho-NF-κB p65 (S529) and nuclear NF-κB p65. DNA fragmentation, YO-PRO-1 staining, mitochondrial depolarization and increased levels of active caspase-3 and cleaved PARP (Asp214) were detected in emetine-treated KG-1a cells. Moreover, treatment with the pancaspase inhibitor Z-VAD(OMe)-FMK partially prevented the apoptotic cell death induced by emetine. Emetine treatment also increased cellular and mitochondrial reactive oxygen species, and emetine-induced apoptosis in KG-1a cells was partially prevented by the antioxidant N-acetylcysteine, indicating that emetine induces apoptosis, at least in part, by inducing oxidative stress. Overall, these studies indicate that emetine is a novel potential anti-AML agent with promising activity against stem/progenitor cells, encouraging the development of further studies aimed at its clinical application.
Collapse
Affiliation(s)
- Suellen L R Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
| | - Ingrid R S B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
| | | | - Rafaela G A Costa
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
| | - Maiara de S Oliveira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
| | - Gabriela A da C Barbosa
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
- SENAI Institute for Innovation in Advanced Health Systems, SENAI CIMATEC, Salvador, BA, 41650-010, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
- Department of Propaedeutics, Federal University of Bahia (UFBA), Salvador, BA, 40301-155, Brazil
| | - Ludmila F Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
| | - Clarissa A G Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
- Department of Propaedeutics, Federal University of Bahia (UFBA), Salvador, BA, 40301-155, Brazil
- Center for Biotechnology and Cell Therapy, D'Or Institute for Research and Education (IDOR), Salvador, BA, 41253-190, Brazil
| | - Nainita Roy
- Department of Pathology, School of Medicine, New York University, New York, NY, 10016, United States of America
| | - Christopher Y Park
- Department of Pathology, School of Medicine, New York University, New York, NY, 10016, United States of America
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil.
| |
Collapse
|
29
|
Roos J, Zinngrebe J, Huber-Lang M, Lupu L, Schmidt MA, Strobel H, Westhoff MA, Stifel U, Gebhard F, Wabitsch M, Mollnes TE, Debatin KM, Halbgebauer R, Fischer-Posovszky P. Trauma-associated extracellular histones mediate inflammation via a MYD88-IRAK1-ERK signaling axis and induce lytic cell death in human adipocytes. Cell Death Dis 2024; 15:285. [PMID: 38653969 PMCID: PMC11039744 DOI: 10.1038/s41419-024-06676-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024]
Abstract
Despite advances in the treatment and care of severe physical injuries, trauma remains one of the main reasons for disability-adjusted life years worldwide. Trauma patients often suffer from disturbances in energy utilization and metabolic dysfunction, including hyperglycemia and increased insulin resistance. White adipose tissue plays an essential role in the regulation of energy homeostasis and is frequently implicated in traumatic injury due to its ubiquitous body distribution but remains poorly studied. Initial triggers of the trauma response are mainly damage-associated molecular patterns (DAMPs) such as histones. We hypothesized that DAMP-induced adipose tissue inflammation contributes to metabolic dysfunction in trauma patients. Therefore, we investigated whether histone release during traumatic injury affects adipose tissue. Making use of a murine polytrauma model with hemorrhagic shock, we found increased serum levels of histones accompanied by an inflammatory response in white adipose tissue. In vitro, extracellular histones induced an inflammatory response in human adipocytes. On the molecular level, this inflammatory response was mediated via a MYD88-IRAK1-ERK signaling axis as demonstrated by pharmacological and genetic inhibition. Histones also induced lytic cell death executed independently of caspases and RIPK1 activity. Importantly, we detected increased histone levels in the bloodstream of patients after polytrauma. Such patients might benefit from a therapy consisting of activated protein C and the FDA-approved ERK inhibitor trametinib, as this combination effectively prevented histone-mediated effects on both, inflammatory gene activation and cell death in adipocytes. Preventing adipose tissue inflammation and adipocyte death in patients with polytrauma could help minimize posttraumatic metabolic dysfunction.
Collapse
Affiliation(s)
- Julian Roos
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Julia Zinngrebe
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Medical Center, Ulm, Germany
| | - Ludmila Lupu
- Institute of Clinical and Experimental Trauma Immunology, University Medical Center, Ulm, Germany
| | - Miriam A Schmidt
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Hannah Strobel
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Ulrich Stifel
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Florian Gebhard
- Department of Orthopedic Trauma, Hand, and Reconstructive Surgery, University Medical Center, Ulm, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Tom Eirik Mollnes
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
- Research Laboratory, Nordland Hospital Trust, Bodo, Norway
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Rebecca Halbgebauer
- Institute of Clinical and Experimental Trauma Immunology, University Medical Center, Ulm, Germany
| | | |
Collapse
|
30
|
Zhuo L, Stöckl JB, Fröhlich T, Moser S, Vollmar AM, Zahler S. A Novel Interaction of Slug (SNAI2) and Nuclear Actin. Cells 2024; 13:696. [PMID: 38667311 PMCID: PMC11049500 DOI: 10.3390/cells13080696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Actin is a protein of central importance to many cellular functions. Its localization and activity are regulated by interactions with a high number of actin-binding proteins. In a yeast two-hybrid (Y2H) screening system, snail family transcriptional repressor 2 (SNAI2 or slug) was identified as a yet unknown potential actin-binding protein. We validated this interaction using immunoprecipitation and analyzed the functional relation between slug and actin. Since both proteins have been reported to be involved in DNA double-strand break (DSB) repair, we focused on their interaction during this process after treatment with doxorubicin or UV irradiation. Confocal microscopy elicits that the overexpression of actin fused to an NLS stabilizes complexes of slug and γH2AX, an early marker of DNA damage repair.
Collapse
Affiliation(s)
- Ling Zhuo
- Center for Drug Research, Ludwig-Maximilians-University Munich, Butenandtstr, 5-13, 81377 Munich, Germany; (L.Z.); (A.M.V.)
| | - Jan B. Stöckl
- Laboratory for Functional Genome Analysis, Gene Center Munich, Ludwig-Maximilians-University Munich, Feodor-Lynen-Str. 25, 81377 Munich, Germany; (J.B.S.); (T.F.)
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis, Gene Center Munich, Ludwig-Maximilians-University Munich, Feodor-Lynen-Str. 25, 81377 Munich, Germany; (J.B.S.); (T.F.)
| | - Simone Moser
- Department of Pharmacognosy, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria;
| | - Angelika M. Vollmar
- Center for Drug Research, Ludwig-Maximilians-University Munich, Butenandtstr, 5-13, 81377 Munich, Germany; (L.Z.); (A.M.V.)
| | - Stefan Zahler
- Center for Drug Research, Ludwig-Maximilians-University Munich, Butenandtstr, 5-13, 81377 Munich, Germany; (L.Z.); (A.M.V.)
| |
Collapse
|
31
|
Habashy KJ, Dmello C, Chen L, Arrieta VA, Kim KS, Gould A, Youngblood MW, Bouchoux G, Burdett KB, Zhang H, Canney M, Stupp R, Sonabend AM. Paclitaxel and Carboplatin in Combination with Low-intensity Pulsed Ultrasound for Glioblastoma. Clin Cancer Res 2024; 30:1619-1629. [PMID: 38295144 DOI: 10.1158/1078-0432.ccr-23-2367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 11/22/2023] [Accepted: 01/29/2024] [Indexed: 02/02/2024]
Abstract
PURPOSE We recently reported on clinical trials for patients with recurrent glioblastoma where low-intensity pulsed ultrasound and microbubbles (LIPU/MB) improved paclitaxel or carboplatin delivery into the brain. Here, we report variable local tumor control with paclitaxel at the maximal/target dose in our phase I trial (NCT04528680). To address this, we investigated the combination of paclitaxel with carboplatin in preclinical glioma models. EXPERIMENTAL DESIGN We performed MRI-based analysis to evaluate disease control in patients from our trial. We studied the cytotoxicity of paclitaxel and carboplatin against 11 human glioma lines as monotherapy and in combination at concentrations derived from human intraoperative studies. Synergy was assessed with the Loewe model and the survival benefit evaluated in two xenografts. We examined the effects on cell cycle progression, DNA damage, and apoptosis. RESULTS Patients treated with paclitaxel and LIPU/MB exhibited variable local tumor control, which correlated with overall survival. We observed limited cross-resistance to paclitaxel and carboplatin in glioma lines, with almost a third of them being exclusively susceptible to one drug. This combination led to susceptibility of 81% of lines and synergy in 55% of them. The combination proved more efficacious in two intracranial xenografts when administered with LIPU/MB, leading to complementary effects on cell cycle arrest. CONCLUSIONS Combining paclitaxel and carboplatin in gliomas may be more efficacious than monotherapy, as in other cancers, due to synergy and independent susceptibility to each drug. These results form the basis for an ongoing phase II trial (NCT04528680) where we investigate this combination with LIPU/MB.
Collapse
Affiliation(s)
- Karl J Habashy
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Crismita Dmello
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Li Chen
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Victor A Arrieta
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Kwang-Soo Kim
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Andrew Gould
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Mark W Youngblood
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | - Kirsten B Burdett
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Hui Zhang
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | - Roger Stupp
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Adam M Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
32
|
Costa RGA, Oliveira MDS, Rodrigues ACBDC, Silva SLR, Dias IRSB, Soares MBP, de Faro Valverde L, Gurgel Rocha CA, Dias RB, Bezerra DP. Bortezomib suppresses acute myelogenous leukaemia stem-like KG-1a cells via NF-κB inhibition and the induction of oxidative stress. J Cell Mol Med 2024; 28:e18333. [PMID: 38652192 PMCID: PMC11037403 DOI: 10.1111/jcmm.18333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/24/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
Acute myelogenous leukaemia (AML) originates and is maintained by leukaemic stem cells (LSCs) that are inherently resistant to antiproliferative therapies, indicating that a critical strategy for overcoming chemoresistance in AML therapy is to eradicate LSCs. In this work, we investigated the anti-AML activity of bortezomib (BTZ), emphasizing its anti-LSC potential, using KG-1a cells, an AML cell line with stem-like properties. BTZ presented potent cytotoxicity to both solid and haematological malignancy cells and reduced the stem-like features of KG-1a cells, as observed by the reduction in CD34- and CD123-positive cells. A reduction in NF-κB p65 nuclear staining was observed in BTZ-treated KG-1a cells, in addition to upregulation of the NF-κB inhibitor gene NFΚBIB. BTZ-induced DNA fragmentation, nuclear condensation, cell shrinkage and loss of transmembrane mitochondrial potential along with an increase in active caspase-3 and cleaved PARP-(Asp 214) level in KG-1a cells. Furthermore, BTZ-induced cell death was partially prevented by pretreatment with the pancaspase inhibitor Z-VAD-(OMe)-FMK, indicating that BTZ induces caspase-mediated apoptosis. BTZ also increased mitochondrial superoxide levels in KG-1a cells, and BTZ-induced apoptosis was partially prevented by pretreatment with the antioxidant N-acetylcysteine, indicating that BTZ induces oxidative stress-mediated apoptosis in KG-1a cells. At a dosage of 0.1 mg/kg every other day for 2 weeks, BTZ significantly reduced the percentage of hCD45-positive cells in the bone marrow and peripheral blood of NSG mice engrafted with KG-1a cells with tolerable toxicity. Taken together, these data indicate that the anti-LSC potential of BTZ appears to be an important strategy for AML treatment.
Collapse
Affiliation(s)
- Rafaela G. A. Costa
- Gonçalo Moniz InstituteOswaldo Cruz Foundation (IGM‐FIOCRUZ/BA)SalvadorBahiaBrazil
| | | | | | - Suellen L. R. Silva
- Gonçalo Moniz InstituteOswaldo Cruz Foundation (IGM‐FIOCRUZ/BA)SalvadorBahiaBrazil
| | - Ingrid R. S. B. Dias
- Gonçalo Moniz InstituteOswaldo Cruz Foundation (IGM‐FIOCRUZ/BA)SalvadorBahiaBrazil
| | - Milena B. P. Soares
- Gonçalo Moniz InstituteOswaldo Cruz Foundation (IGM‐FIOCRUZ/BA)SalvadorBahiaBrazil
- SENAI Institute for Innovation in Advanced Health SystemsSENAI CIMATECSalvadorBahiaBrazil
| | | | - Clarissa Araujo Gurgel Rocha
- Gonçalo Moniz InstituteOswaldo Cruz Foundation (IGM‐FIOCRUZ/BA)SalvadorBahiaBrazil
- Department of Propaedeutics, Faculty of DentistryFederal University of Bahia (UFBA)SalvadorBahiaBrazil
- Center for Biotechnology and Cell TherapyD'Or Institute for Research and Education (IDOR)SalvadorBahiaBrazil
| | - Rosane Borges Dias
- Gonçalo Moniz InstituteOswaldo Cruz Foundation (IGM‐FIOCRUZ/BA)SalvadorBahiaBrazil
- Department of Propaedeutics, Faculty of DentistryFederal University of Bahia (UFBA)SalvadorBahiaBrazil
| | - Daniel P. Bezerra
- Gonçalo Moniz InstituteOswaldo Cruz Foundation (IGM‐FIOCRUZ/BA)SalvadorBahiaBrazil
| |
Collapse
|
33
|
Ibrahim BT, Allam HA, El-Dydamony NM, Fouad MA, Mohammed ER. Exploring new quinazolin-4(3H)-one derivatives as CDK2 inhibitors: Design, synthesis, and anticancer evaluation. Drug Dev Res 2024; 85:e22163. [PMID: 38419305 DOI: 10.1002/ddr.22163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/13/2024] [Accepted: 02/10/2024] [Indexed: 03/02/2024]
Abstract
In the present work, five series of new 2,3-disubstituted quinazolin-4(3H)-ones 4a-c, 5a-d, 6a-g, 7a,b, and 9a-c were designed, synthesized, and screened in vitro for their cytotoxic activity against 60 cancer cell lines by the National Cancer Institute, USA. Five candidates 4c, 6a, 6b, 6d, and 6g revealed promising cytotoxicity with significant percentage growth inhibition in the range of 81.98%-96.45% against the central nervous system (CNS) (SNB-19), melanoma (MDA-MB-435), and non-small cell lung cancer (HOP-62) cell lines. The in vitro cytotoxic half maximal inhibitory concentration (IC50 ) values for the most active compounds 4c, 6a, 6b, 6d, and 6g against the most sensitive cell lines were evaluated. Additionally, screening their cyclin-dependent kinase 2 (CDK2) inhibitory activity was performed. Ortho-chloro-benzylideneamino derivative 6b emerged as the most potent compound with IC50 = 0.67 µM compared to Roscovitine (IC50 = 0.64 µM). The most active candidates arrested the cell cycle at G1, S phases, or both, leading to cell death and inducing apoptosis against CNS (SNB-19), melanoma (MDA-MB-435), and non-small cell lung cancer (HOP-62) cell lines. The molecular docking study verified the resulting outcomes for the most active candidates in the CDK2-binding pocket. Finally, physicochemical, and pharmacokinetic properties deduced that compounds 4c, 6a, 6b, 6d, and 6g displayed significant drug-likeness properties. According to the obtained results, the newly targeted compounds are regarded as promising scaffolds for the continued development of novel CDK2 inhibitors.
Collapse
Affiliation(s)
- Basant T Ibrahim
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Egypt
| | | | - Nehad M El-Dydamony
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Egypt
| | - Marwa A Fouad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Pharmaceutical Chemistry Department, New Giza University, Cairo, Egypt
| | - Eman R Mohammed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
34
|
Rodrigues ACBDC, Silva SLR, Dias IRSB, Costa RGA, Oliveira MDS, Soares MBP, Dias RB, Valverde LF, Rocha CAG, Johnson EM, Pina C, Bezerra DP. Piplartine eliminates CD34 + AML stem/progenitor cells by inducing oxidative stress and suppressing NF-κB signalling. Cell Death Discov 2024; 10:147. [PMID: 38503729 PMCID: PMC10951277 DOI: 10.1038/s41420-024-01909-4] [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: 01/11/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/21/2024] Open
Abstract
Acute myeloid leukaemia (AML) is a haematological malignancy characterised by the accumulation of transformed myeloid progenitors in the bone marrow. Piplartine (PL), also known as piperlongumine, is a pro-oxidant small molecule extracted from peppers that has demonstrated antineoplastic potential in solid tumours and other haematological malignancies. In this work, we explored the potential of PL to treat AML through the use of a combination of cellular and molecular analyses of primary and cultured leukaemia cells in vitro and in vivo. We showed that PL exhibits in vitro cytotoxicity against AML cells, including CD34+ leukaemia-propagating cells, but not healthy haematopoietic progenitors, suggesting anti-leukaemia selectivity. Mechanistically, PL treatment increased reactive oxygen species (ROS) levels and induced ROS-mediated apoptosis in AML cells, which could be prevented by treatment with the antioxidant scavenger N-acetyl-cysteine and the pancaspase inhibitor Z-VAD(OMe)-FMK. PL treatment reduced NFKB1 gene transcription and the level of NF-κB p65 (pS536), which was depleted from the nucleus of AML cells, indicating suppression of NF-κB p65 signalling. Significantly, PL suppressed AML development in a mouse xenograft model, and its combination with current AML treatments (cytarabine, daunorubicin and azacytidine) had synergistic effects, indicating translational therapeutic potential. Taken together, these data position PL as a novel anti-AML candidate drug that can target leukaemia stem/progenitors and is amenable to combinatorial therapeutic strategies.
Collapse
Affiliation(s)
- Ana Carolina B da C Rodrigues
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Suellen L R Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Ingrid R S B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Rafaela G A Costa
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Maiara de S Oliveira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
- SENAI Institute for Innovation in Advanced Health Systems, SENAI CIMATEC, Salvador, Bahia, 41650-010, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
- Department of Propaedeutics and Integrated Clinical, Faculty of Dentistry, Federal University of Bahia (UFBA), Salvador, Bahia, 40301-155, Brazil
| | - Ludmila F Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Clarissa A G Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
- Department of Propaedeutics and Integrated Clinical, Faculty of Dentistry, Federal University of Bahia (UFBA), Salvador, Bahia, 40301-155, Brazil
- Center for Biotechnology and Cell Therapy, D'Or Institute for Research and Education (IDOR), Salvador, Bahia, 41253-190, Brazil
| | - Emily M Johnson
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Cristina Pina
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK.
- Centre for Genome Engineering and Maintenance, Brunel University London, Uxbridge, UB8 3PH, UK.
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil.
| |
Collapse
|
35
|
Schmitt L, Lechtenberg I, Drießen D, Flores-Romero H, Skowron MA, Sekeres M, Hoppe J, Krings KS, Llewellyn TR, Peter C, Stork B, Qin N, Bhatia S, Nettersheim D, Fritz G, García-Sáez AJ, Müller TJJ, Wesselborg S. Novel meriolin derivatives activate the mitochondrial apoptosis pathway in the presence of antiapoptotic Bcl-2. Cell Death Discov 2024; 10:125. [PMID: 38461295 PMCID: PMC10924942 DOI: 10.1038/s41420-024-01901-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/21/2024] [Accepted: 02/29/2024] [Indexed: 03/11/2024] Open
Abstract
Meriolin derivatives represent a new class of kinase inhibitors with a pronounced cytotoxic potential. Here, we investigated a newly synthesized meriolin derivative (termed meriolin 16) that displayed a strong apoptotic potential in Jurkat leukemia and Ramos lymphoma cells. Meriolin 16 induced apoptosis in rapid kinetics (within 2-3 h) and more potently (IC50: 50 nM) than the previously described derivatives meriolin 31 and 36 [1]. Exposure of Ramos cells to meriolin 16, 31, or 36 for 5 min was sufficient to trigger severe and irreversible cytotoxicity. Apoptosis induction by all three meriolin derivatives was independent of death receptor signaling but required caspase-9 and Apaf-1 as central mediators of the mitochondrial death pathway. Meriolin-induced mitochondrial toxicity was demonstrated by disruption of the mitochondrial membrane potential (ΔΨm), mitochondrial release of proapoptotic Smac, processing of the dynamin-like GTPase OPA1, and subsequent fragmentation of mitochondria. Remarkably, all meriolin derivatives were able to activate the mitochondrial death pathway in Jurkat cells, even in the presence of the antiapoptotic Bcl-2 protein. In addition, meriolins were capable of inducing cell death in imatinib-resistant K562 and KCL22 chronic myeloid leukemia cells as well as in cisplatin-resistant J82 urothelial carcinoma and 2102EP germ cell tumor cells. Given the frequent inactivation of the mitochondrial apoptosis pathway by tumor cells, such as through overexpression of antiapoptotic Bcl-2, meriolin derivatives emerge as promising therapeutic agents for overcoming treatment resistance.
Collapse
Affiliation(s)
- Laura Schmitt
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Ilka Lechtenberg
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Daniel Drießen
- Institute of Organic Chemistry and Macromolecular Chemistry, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Hector Flores-Romero
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, 50931, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
- Ikerbasque, Basque Foundation for Science, 48013, Bilbao, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Margaretha A Skowron
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, Düsseldorf, Germany
| | - Marlena Sekeres
- Institute of Toxicology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Julia Hoppe
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Karina S Krings
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Tanya R Llewellyn
- Clinic of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Christoph Peter
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Björn Stork
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Nan Qin
- Clinic of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Sanil Bhatia
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Daniel Nettersheim
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, Düsseldorf, Germany
| | - Gerhard Fritz
- Institute of Toxicology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Ana J García-Sáez
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, 50931, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
| | - Thomas J J Müller
- Institute of Organic Chemistry and Macromolecular Chemistry, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Sebastian Wesselborg
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany.
| |
Collapse
|
36
|
Serag MI, Tawfik SS, Badr SMI, Eisa HM. New oxadiazole and pyrazoline derivatives as anti-proliferative agents targeting EGFR-TK: design, synthesis, biological evaluation and molecular docking study. Sci Rep 2024; 14:5474. [PMID: 38443456 PMCID: PMC10915170 DOI: 10.1038/s41598-024-55046-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 02/20/2024] [Indexed: 03/07/2024] Open
Abstract
Two new series of oxadiazole and pyrazoline derivatives were designed and synthesized as promising EGFR-TK inhibitors. The in vitro antiproliferative activity was studied against three human cancer cell lines; HCT116, HepG-2 and MCF7 using MTT assay. Compound 10c showed the most potent anticancer activity against all cancer cell lines, with IC50 range of 1.82 to 5.55 μM, while proving safe towards normal cells WI-38 (IC50 = 41.17 μM) compared to the reference drug doxorubicin (IC50 = 6.72 μM). The most active candidates 5a, 9b, 10a, 10b and 10c were further assessed for their EGFR-TK inhibition. The best of which, compounds 5a and 10b showed IC50 of 0.09 and 0.16 μM respectively compared to gefitinib (IC50 = 0.04 μM). Further investigation against other EGFR family members, showed that 5a displayed good activities against HER3 and HER4 with IC50 values 0.18 and 0.37 µM, respectively compared to gefitinib (IC50 = 0.35 and 0.58 µM, respectively). Furthermore, 5a was evaluated for cell cycle distribution and apoptotic induction on HepG-2 cells. It induced mitochondrial apoptotic pathway and increased accumulation of ROS. Molecular docking study came in agreement with the biological results. Compounds 5a and 10b showed promising drug-likeness with good physicochemical properties.
Collapse
Affiliation(s)
- Marwa I Serag
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Samar S Tawfik
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Sahar M I Badr
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Hassan M Eisa
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
37
|
Kang X, Chen H, Zhou Z, Tu S, Cui B, Li Y, Dong S, Zhang Q, Xu Y. Targeting Cyclin-Dependent Kinase 1 Induces Apoptosis and Cell Cycle Arrest of Activated Hepatic Stellate Cells. Adv Biol (Weinh) 2024; 8:e2300403. [PMID: 38103005 DOI: 10.1002/adbi.202300403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/08/2023] [Indexed: 12/17/2023]
Abstract
Liver fibrosis is the integral process of chronic liver diseases caused by multiple etiologies and characterized by excessive deposition of extracellular matrix (ECM). During liver fibrosis, hepatic stellate cells (HSCs) transform into a highly proliferative, activated state, producing various cytokines, chemokines, and ECM. However, the precise mechanisms that license HSCs into the highly proliferative state remain unclear. Cyclin-dependent kinase 1 (CDK1) is a requisite event for the transition of the G1/S and G2/M phases in eukaryotic cells. In this study, it is demonstrated that CDK1 and its activating partners, Cyclin A2 and Cyclin B1, are upregulated in both liver fibrosis/cirrhosis patient specimens and the murine hepatic fibrosis models, especially in activated HSCs. In vitro, CDK1 is upregulated in spontaneously activated HSCs, and inhibiting CDK1 with specific small-molecule inhibitors (CGP74514A, RO-3306, or Purvalanol A) orshort hairpin RNAs (shRNAs) resulted in HSC apoptosis and cell cycle arrest by regulating Survivin expression. Above all, it is illustrated that increased CDK1 expression licenses the HSCs into a highly proliferative state and can serve as a potential therapeutic target in liver fibrosis.
Collapse
Affiliation(s)
- Xinmei Kang
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Huaxin Chen
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Zhuowei Zhou
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Silin Tu
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Bo Cui
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Yanli Li
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Shuai Dong
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Qi Zhang
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
- Cell-gene Therapy Translational Medicine Research Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Yan Xu
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| |
Collapse
|
38
|
Patterson C, Hazime KS, Zelenay S, Davis DM. Prostaglandin E₂ impacts multiple stages of the natural killer cell antitumor immune response. Eur J Immunol 2024; 54:e2350635. [PMID: 38059519 DOI: 10.1002/eji.202350635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/08/2023]
Abstract
Tumor immune escape is a major factor contributing to cancer progression and unresponsiveness to cancer therapies. Tumors can produce prostaglandin E2 (PGE2 ), an inflammatory mediator that directly acts on Natural killer (NK) cells to inhibit antitumor immunity. However, precisely how PGE2 influences NK cell tumor-restraining functions remains unclear. Here, we report that following PGE₂ treatment, human NK cells exhibited altered expression of specific activating receptors and a reduced ability to degranulate and kill cancer targets. Transcriptional analysis uncovered that PGE₂ also differentially modulated the expression of chemokine receptors by NK cells, inhibiting CXCR3 but increasing CXCR4. Consistent with this, PGE₂-treated NK cells exhibited decreased migration to CXCL10 but increased ability to migrate toward CXCL12. Using live cell imaging, we showed that in the presence of PGE2 , NK cells were slower and less likely to kill cancer target cells following conjugation. Imaging the sequential stages of NK cell killing revealed that PGE₂ impaired NK cell polarization, but not the re-organization of synaptic actin or the release of perforin itself. Together, these findings demonstrate that PGE₂ affects multiple but select NK cell functions. Understanding how cancer cells subvert NK cells is necessary to more effectively harness the cancer-inhibitory function of NK cells in treatments.
Collapse
Affiliation(s)
- Chloe Patterson
- The Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, United Kingdom
| | - Khodor S Hazime
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, South Kensington, London, United Kingdom
| | - Santiago Zelenay
- The Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, United Kingdom
- Cancer Inflammation and Immunity Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Daniel M Davis
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, South Kensington, London, United Kingdom
| |
Collapse
|
39
|
Fettucciari K, Spaterna A, Marconi P, Bassotti G. Pro-Inflammatory Cytokines Enhanced In Vitro Cytotoxic Activity of Clostridioides difficile Toxin B in Enteric Glial Cells: The Achilles Heel of Clostridioides difficile Infection? Int J Mol Sci 2024; 25:958. [PMID: 38256032 PMCID: PMC10815653 DOI: 10.3390/ijms25020958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Bacterial infections are characterized by an inflammatory response, which is essential for infection containment but is also responsible for negative effects on the host. The pathogen itself may have evolved molecular mechanisms to antagonize the antimicrobial effects of an inflammatory response and to enhance its pathogenicity using inflammatory response mediators, such as cytokines. Clostridioides difficile (C. difficile) infection (CDI) causes gastrointestinal diseases with markedly increasing global incidence and mortality rates. The main C. difficile virulence factors, toxin A and B (TcdA/TcdB), cause cytopathic/cytotoxic effects and inflammation. We previously demonstrated that TcdB induces enteric glial cell (EGC) apoptosis, which is enhanced by the pro-inflammatory cytokine tumor necrosis factor alpha plus interferon gamma (CKs). However, it is unknown whether CKs-enhanced TcdB cytotoxicity (apoptosis/necrosis) is affected by the timing of the appearance of the CKs. Thus, we simulated in vitro, in our experimental model with TcdB and EGCs, three main situations of possible interactions between TcdB and the timing of CK stimulation: before TcdB infection, concomitantly with infection, or at different times after infection and persisting over time. In these experimental conditions, which all represent situations of possible interactions between C. difficile and the timing of CK stimulation, we evaluated apoptosis, necrosis, and cell cycle phases. The CKs, in all of these conditions, enhanced TcdB cytotoxicity, which from apoptosis became necrosis when CK stimulation persisted over time, and was most relevant after 48 h of TcdB:EGCs interaction. Particularly, the enhancement of apoptosis by CKs was dependent on the TcdB dose and in a less relevant manner on the CK stimulation time, while the enhancement of necrosis occurred always independently of the TcdB dose and CK stimulation time. However, since in all conditions stimulation with CKs strongly enhanced the TcdB cytotoxicity, it always had a negative impact on C. difficile pathogenicity. This study might have important implications for the treatment of CDI.
Collapse
Affiliation(s)
- Katia Fettucciari
- Biosciences & Medical Embryology Section, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy;
| | - Andrea Spaterna
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Macerata, Italy
| | - Pierfrancesco Marconi
- Biosciences & Medical Embryology Section, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy;
| | - Gabrio Bassotti
- Gastroenterology, Hepatology & Digestive Endoscopy Section, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy;
- Gastroenterology & Hepatology Unit, Santa Maria Della Misericordia Hospital, 06129 Perugia, Italy
| |
Collapse
|
40
|
Rostampour S, Eslami F, Babaei E, Mostafavi H, Mahdavi M. An Active Compound from the Pyrazine Family Induces Apoptosis by Targeting the Bax/Bcl2 and Survivin Expression in Chronic Myeloid Leukemia K562 Cells. Anticancer Agents Med Chem 2024; 24:203-212. [PMID: 38038011 DOI: 10.2174/0118715206272359231121105713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/03/2023] [Accepted: 10/19/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND It has been established that pyrazine derivatives, which have widespread bioactivities, can effectively treat cancer. OBJECTIVES In this study, we investigated the effects of 2-methoxy-5-(oxiran-2-ylmethyl) phenyl pyrazine-2- carboxylate (2-mOPP), a new pyrazine derivative, on proliferation, viability, and apoptosis induction in human leukemia K562 cells. METHODS For this purpose, the K562 cells were treated with various concentrations (20-120 μM) of the 2-mOPP for 24-72 hours. Cell viability was determined by MTT growth inhibition assay. Apoptotic activity of 2-mOPP was investigated morphologically by Hoechst staining, cell surface expression assay of phosphatidylserine by Annexin-V/PI technique, as well as DNA fragmentation assay. The effect of 2-mOPP on the K562 cell cycle was studied by flow cytometry. To determine the impact of 2-mOPP on the expression of intrinsic apoptosis-related genes, Bcl2 (anti-apoptotic), Bax (pro-apoptotic), and Survivin genes expression levels were evaluated before and after treatment with 2-mOPP through Real-Time PCR analysis. RESULTS The results revealed that 2-mOPP inhibited viability with IC50 of 25μM in 72 h. Morphological changes assessment by fluorescence microscopy, Annexin V/PI double staining by flow cytometry, and DNA ladders formation upon cell treatment with the 2-mOPP showed that this compound induces apoptosis at IC50 value. Cell cycle arrest was observed in the G0/G1 phase, and the sub-G1 cell population (the sign of apoptosis) increased in a time-dependent manner. Low expression levels of Bcl2 and Survivin in K562 cells were observed 24-72 h after treatment. Along with the down-regulation of Survivin and Bcl2, the expression of Bax was increased after treatment with 2-mOPP. CONCLUSION These findings demonstrate that the new pyrazine derivative plays a crucial role in blocking the proliferation of the leukemic cells by inducing cell cycle arrest and apoptosis.
Collapse
Affiliation(s)
- Saeedeh Rostampour
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Farhad Eslami
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Esmaeil Babaei
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Hossein Mostafavi
- Department of Organic Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Majid Mahdavi
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| |
Collapse
|
41
|
van Zyl E, Peneycad C, Perehiniak E, McKay BC. Cyclin-dependent kinase inhibitor 1 plays a more prominent role than activating transcription factor 4 or the p53 tumour suppressor in thapsigargin-induced G1 arrest. PeerJ 2023; 11:e16683. [PMID: 38130926 PMCID: PMC10734451 DOI: 10.7717/peerj.16683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
Background Thapsigargin (Tg) is a compound that inhibits the SERCA calcium transporter leading to decreased endoplasmic reticulum (ER) Ca2+ levels. Many ER chaperones are required for proper folding of membrane-associated and secreted proteins, and they are Ca2+ dependent. Therefore, Tg leads to the accumulation of misfolded proteins in the ER, activating the unfolded protein response (UPR) to help restore homeostasis. Tg reportedly induces cell cycle arrest and apoptosis in many cell types but how these changes are linked to the UPR remains unclear. The activating transcription factor 4 (ATF4) plays a key role in regulating ER stress-induced gene expression so we sought to determine if ATF4 is required for Tg-induced cell cycle arrest and apoptosis using ATF4-deficient cells. Methods Two-parameter flow cytometric analysis of DNA replication and DNA content was used to assess the effects of Tg on cell cycle distribution in isogenic HCT116-derived cell lines either expressing or lacking ATF4. For comparison, we similarly assessed the Tg response in isogenic cell lines deleted of the p53 tumour suppressor and the p53-regulated p21WAF1 cyclin-dependent kinase inhibitor important in G1 and G2 arrests induced by DNA damage. Results Tg led to a large depletion of the S phase population with a prominent increase in the proportion of HCT116 cells in the G1 phase of the cell cycle. Importantly, this effect was largely independent of ATF4. We found that loss of p21WAF1 but not p53 permitted Tg treated cells to enter S phase and synthesize DNA. Therefore, p21WAF1plays an important role in these Tg-induced cell cycle alterations while ATF4 and p53 do not. Remarkably, the ATF4-, p53-and p21WAF1-deficient cell lines were all more sensitive to Tg-induced apoptosis. Taken together, p21WAF1 plays a larger role in regulating Tg-induced G1 and G2 arrests than ATF4 or p53 but these proteins similarly contribute to protection from Tg-induced apoptosis. This work highlights the complex network of stress responses that are activated in response to ER stress.
Collapse
Affiliation(s)
- Erin van Zyl
- Department of Biology, Carleton University, Ottawa, ON, Canada
| | - Claire Peneycad
- Department of Biology, Carleton University, Ottawa, ON, Canada
| | - Evan Perehiniak
- Department of Biology, Carleton University, Ottawa, ON, Canada
| | - Bruce C. McKay
- Department of Biology, Carleton University, Ottawa, ON, Canada
- Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
42
|
Silva VR, Santos LDS, de Castro MVL, Dias RB, Valverde LDF, Rocha CAG, Soares MBP, Quadros CA, Correa RS, Batista AA, Bezerra DP. A novel ruthenium complex with 5-fluorouracil suppresses colorectal cancer stem cells by inhibiting Akt/mTOR signaling. Cell Death Discov 2023; 9:460. [PMID: 38104089 PMCID: PMC10725484 DOI: 10.1038/s41420-023-01759-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 12/19/2023] Open
Abstract
[Ru(5-FU)(PPh3)2(bipy)]PF6 (Ru/5-FU) is a novel ruthenium complex with 5-fluorouracil with promising potential against colorectal cancer (CRC). In the present study, we investigated the molecular mechanism of Ru/5-FU action in HCT116 CRC cells. Ru/5-FU exhibited potent cytotoxicity on a panel of cancer cell lines and on primary cancer cells and induced apoptosis in HCT116 CRC cells. Ru/5-FU reduced AKT1 gene transcripts, as well as the expression of Akt1 and Akt (pS473) and downstream Akt proteins mTOR (pS2448), S6 (pS235/pS236), 4EBP1 (pT36/pT45), GSK-3β (pS9) and NF-κB p65 (pS529), but not Akt upstream proteins Hsp90 and PI3K p85/p55 (pT458/pT199), indicating an inhibitory action of Akt/mTOR signaling. Ru/5-FU increased LC3B expression and reduced p62/SQSTM1 levels, indicating autophagy induction. Curiously, the autophagy inhibitors 3-methyladenine and chloroquine increased Ru/5-FU-induced cell death, indicating an induction of cytoprotective autophagy by this compound. Ru/5-FU also reduced clonogenic survival, as well as the percentage of CD133+ cells and colonosphere formation, indicating that Ru/5-FU can suppress stem cells in HCT116 cells. Ru/5-FU inhibited cell migration and invasion in wound healing assays and Transwell cell invasion assays, along with a reduction in vimentin expression and an increase in E-cadherin levels, indicating that Ru/5-FU can interfere with epithelial-mesenchymal transition. Ru/5-FU also inhibited in vivo HCT116 cell development and experimental lung metastases in mouse xenograft models. Altogether, these results indicate that Ru/5-FU is an anti-CRC chemotherapy drug candidate with the ability to suppress stemness in CRC cells by inhibiting Akt/mTOR signaling.
Collapse
Affiliation(s)
- Valdenizia R Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Luciano de S Santos
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Maria V L de Castro
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador, Bahia, 40110-909, Brazil
| | - Ludmila de F Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Clarissa A G Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador, Bahia, 40110-909, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
- SENAI Institute of Innovation (ISI) in Health Advanced Systems, University Center SENAI/CIMATEC, Salvador, Bahia, 41650-010, Brazil
| | - Claudio A Quadros
- São Rafael Hospital, Rede D'Or/São Luiz, Salvador, Bahia, 41253-190, Brazil
- Bahia State University, Salvador, Bahia, 41150-000, Brazil
| | - Rodrigo S Correa
- Department of Chemistry, Federal University of Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Alzir A Batista
- Department of Chemistry, Federal University of São Carlos, São Carlos, São Paulo, 13561-901, Brazil
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil.
| |
Collapse
|
43
|
Santos LDS, Silva VR, de Castro MVL, Dias RB, Valverde LDF, Rocha CAG, Soares MBP, Quadros CA, Dos Santos ER, Oliveira RMM, Carlos RM, Nogueira PCL, Bezerra DP. New ruthenium-xanthoxylin complex eliminates colorectal cancer stem cells by targeting the heat shock protein 90 chaperone. Cell Death Dis 2023; 14:832. [PMID: 38102125 PMCID: PMC10724293 DOI: 10.1038/s41419-023-06330-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 11/14/2023] [Accepted: 11/23/2023] [Indexed: 12/17/2023]
Abstract
In this work, we describe a novel ruthenium-xanthoxylin complex, [Ru(phen)2(xant)](PF6) (RXC), that can eliminate colorectal cancer (CRC) stem cells by targeting the chaperone Hsp90. RXC exhibits potent cytotoxicity in cancer cell lines and primary cancer cells, causing apoptosis in HCT116 CRC cells, as observed by cell morphology, YO-PRO-1/PI staining, internucleosomal DNA fragmentation, mitochondrial depolarization, and PARP cleavage (Asp214). Additionally, RXC can downregulate the HSP90AA1 and HSP90B1 genes and the expression of HSP90 protein, as well as the expression levels of its downstream/client elements Akt1, Akt (pS473), mTOR (pS2448), 4EBP1 (pT36/pT45), GSK-3β (pS9), and NF-κB p65 (pS529), implying that these molecular chaperones can be molecular targets for RXC. Moreover, this compound inhibited clonogenic survival, the percentage of the CRC stem cell subpopulation, and colonosphere formation, indicating that RXC can eliminate CRC stem cells. RXC reduced cell migration and invasion, decreased vimentin and increased E-cadherin expression, and induced an autophagic process that appeared to be cytoprotective, as autophagy inhibitors enhanced RXC-induced cell death. In vivo studies showed that RXC inhibits tumor progression and experimental metastasis in mice with CRC HCT116 cell xenografts. Taken together, these results highlight the potential of the ruthenium complex RXC in CRC therapy with the ability to eliminate CRC stem cells by targeting the chaperone Hsp90.
Collapse
Affiliation(s)
- Luciano de S Santos
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
| | - Valdenizia R Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
| | - Maria V L de Castro
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador, BA, 40110-909, Brazil
| | - Ludmila de F Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
| | - Clarissa A G Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador, BA, 40110-909, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil
- SENAI Institute of Innovation (ISI) in Health Advanced Systems, University Center SENAI/CIMATEC, Salvador, BA, 41650-010, Brazil
| | - Claudio A Quadros
- São Rafael Hospital, Rede D'Or/São Luiz, Salvador, BA, 41253-190, Brazil
- Bahia State University, Salvador, BA, 41150-000, Brazil
| | - Edjane R Dos Santos
- Institute of Natural, Human and Social Sciences, Federal University of Mato Grosso, Sinop, MT, 78557-267, Brazil
| | - Regina M M Oliveira
- Coordination of Science and Technology, Balsas Science Center, Federal University of Maranhão, Balsas, MA, 65800-000, Brazil
| | - Rose M Carlos
- Department of Chemistry, Federal University of São Carlos, São Carlos, SP, 13561-901, Brazil
| | - Paulo C L Nogueira
- Department of Chemistry, Federal University of Sergipe, São Cristóvão, SE, 49100-000, Brazil
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA, 40296-710, Brazil.
| |
Collapse
|
44
|
Panagiotidou C, Burgers LD, Tsadila C, Almpani C, Krigas N, Mossialos D, Rallis MC, Fürst R, Karioti A. HPLC- and NMR-Based Chemical Profiling, Wound-Healing Potential, Anti-Inflammatory and Antibacterial Activities of Satureja pilosa (Lamiaceae), a Neglected Medicinal-Aromatic Herb. PLANTS (BASEL, SWITZERLAND) 2023; 12:4114. [PMID: 38140440 PMCID: PMC10747026 DOI: 10.3390/plants12244114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023]
Abstract
Satureja pilosa Velen. (Lamiaceae) is a perennial and melliferous aromatic-medicinal subshrub which is range-restricted in adjacent parts of Greece and Bulgaria and locally in Italy, known in Northern Greece as wild oregano ("agriorigani") and traditionally collected from the wild for culinary purposes. Since the ethnopharmacological data and modern biological activities of Satureja spp. suggest promising applications in skin conditions, the present study aimed to investigate the hitherto unknown phenolic content of cultivated S. pilosa and its potential biological activities, focusing mainly on wound-healing and anti-inflammatory effects. An HPLC-PDA-MS-targeted phytochemical investigation, along with NMR, allowed for the isolation and characterization of the main constituents, resulting in 18 compounds. Representative extracts and purified compounds were tested for wound-healing activity on NIH/3T3 fibroblasts. The butanol extract exhibited a significantly higher cell migration rate (73.4%) compared to aqueous (50.6%) and methanolic (49.6%) ones, enhancing the cell migration more rapidly at both concentration levels, whilst rosmarinic acid was the most potent among the isolated compounds, with a migration rate of 64.0% at the concentration level of 10-5 mg/mL, followed by 3,4-dihydrophenyllactic acid (54.7%). Moreover, potential effects on endothelial activation processes were explored, including the leukocyte-endothelial cell interaction during inflammatory processes and the migratory capacity during angiogenic actions, since these processes are commonly associated with skin diseases. Finally, extracts and purified compounds demonstrated weak antibacterial potential against two important pathogens (Staphylococcus aureus and Pseudomonas aeruginosa), suggesting that further investigation is warrented.
Collapse
Affiliation(s)
- Christina Panagiotidou
- Laboratory of Pharmacognosy, School of Pharmacy, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece;
| | - Luisa D. Burgers
- Institute of Pharmaceutical Biology, Faculty of Biochemistry, Chemistry and Pharmacy, Goethe University, 60438 Frankfurt, Germany; (L.D.B.); (R.F.)
| | - Christina Tsadila
- Microbial Biotechnology-Molecular Bacteriology-Virology Laboratory, Department of Biochemistry & Biotechnology, University of Thessaly, 41500 Larissa, Greece; (C.T.); (D.M.)
| | - Chara Almpani
- Department of Pharmaceutical Technology, School of Pharmacy, National & Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15784 Athens, Greece; (C.A.); (M.C.R.)
| | - Nikos Krigas
- Hellenic Agricultural Organization—Demeter (ELGO DIMITRA), Institute of Breeding and Plant Genetic Resources, 57001 Thermi, Greece;
| | - Dimitris Mossialos
- Microbial Biotechnology-Molecular Bacteriology-Virology Laboratory, Department of Biochemistry & Biotechnology, University of Thessaly, 41500 Larissa, Greece; (C.T.); (D.M.)
| | - Michail Christou Rallis
- Department of Pharmaceutical Technology, School of Pharmacy, National & Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15784 Athens, Greece; (C.A.); (M.C.R.)
| | - Robert Fürst
- Institute of Pharmaceutical Biology, Faculty of Biochemistry, Chemistry and Pharmacy, Goethe University, 60438 Frankfurt, Germany; (L.D.B.); (R.F.)
- LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), 60325 Frankfurt, Germany
| | - Anastasia Karioti
- Laboratory of Pharmacognosy, School of Pharmacy, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece;
| |
Collapse
|
45
|
Gheybi F, Rajabian F, Tayarani-Najaran Z, Adibi AR, Alavizadeh SH, Kesharwani P, Sahebkar A. Liposomal silymarin anti-oxidative and anti-apoptotic features in lung cells: An implication in cadmium toxicity. J Trace Elem Med Biol 2023; 80:127291. [PMID: 37672872 DOI: 10.1016/j.jtemb.2023.127291] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/25/2023] [Accepted: 08/21/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND Several metallic elements with high atomic weight and density are serious systemic toxicants, and their wide environmental distribution increase the risk of their exposure to human. Silymarin (SL), a polyphenol from milk thistle (Silybum marianum) plant has shown protective role against heavy metal toxicity. However, its low aqueous solubility and rapid metabolism limits its therapeutic potential in clinic. METHODS We compared the role of silymarin nanoliposomes (SL-L) against cadmium (Cd) toxicity in normal MRC-5 and A 549 cancer cells. MRC-5 and A 549 cells exposed to Cd at 25 and 0.25 µM respectively, were treated with various non-toxic SL-L concentrations (2.5, 5, 10 µM) and cells viability, reactive oxygen species (ROS) generation, apoptosis and levels of cleaved PARP and caspase-3 proteins were determined following incubation. RESULTS Results indicated that Cd exposure significantly increased apoptosis due to ROS generation, and showed greater toxicity on cancer cells compared to normal cells. While SL-L at higher concentrations (25 µM and higher) exhibits pro-apoptotic features, lower concentrations (10 and 2.5 µM for MRC-5 and A 549 cancer cells, respectively) played a protective and anti-oxidant role in Cd induced toxicity in both cells. Further, lower SL-L was required to protect cancer cells against Cd toxicity. In general, treatment with SL-L significantly improved cell survival by decreasing ROS levels, cleaved PARP and caspase-3 in both MRC-5 and A 549 cells compared to free silymarin. CONCLUSION Results demonstrated that SL-L potential in protecting against Cd-induced toxicity depends on concentration-dependent antioxidant and anti-apoptotic balance.
Collapse
Affiliation(s)
- Fatemeh Gheybi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Rajabian
- Department of Pharmacodynamics and Toxicology, School of Pharmacy; Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Tayarani-Najaran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Toxicology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Reza Adibi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy; Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India; Center for Global health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
46
|
Shokouhi AR, Chen Y, Yoh HZ, Brenker J, Alan T, Murayama T, Suu K, Morikawa Y, Voelcker NH, Elnathan R. Engineering Efficient CAR-T Cells via Electroactive Nanoinjection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304122. [PMID: 37434421 DOI: 10.1002/adma.202304122] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/13/2023]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has emerged as a promising cell-based immunotherapy approach for treating blood disorders and cancers, but genetically engineering CAR-T cells is challenging due to primary T cells' sensitivity to conventional gene delivery approaches. The current viral-based method can typically involve significant operating costs and biosafety hurdles, while bulk electroporation (BEP) can lead to poor cell viability and functionality. Here, a non-viral electroactive nanoinjection (ENI) platform is developed to efficiently negotiate the plasma membrane of primary human T cells via vertically configured electroactive nanotubes, enabling efficient delivery (68.7%) and expression (43.3%) of CAR genes in the T cells, with minimal cellular perturbation (>90% cell viability). Compared to conventional BEP, the ENI platform achieves an almost threefold higher CAR transfection efficiency, indicated by the significantly higher reporter GFP expression (43.3% compared to 16.3%). By co-culturing with target lymphoma Raji cells, the ENI-transfected CAR-T cells' ability to effectively suppress lymphoma cell growth (86.9% cytotoxicity) is proved. Taken together, the results demonstrate the platform's remarkable capacity to generate functional and effective anti-lymphoma CAR-T cells. Given the growing potential of cell-based immunotherapies, such a platform holds great promise for ex vivo cell engineering, especially in CAR-T cell therapy.
Collapse
Affiliation(s)
- Ali-Reza Shokouhi
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Hao Zhe Yoh
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Jason Brenker
- Dynamic Micro Devices (DMD) Lab, Department of Mechanical & Aerospace Engineering, Monash University, 17 College Walk, Clayton, VIC, 3168, Australia
| | - Tuncay Alan
- Dynamic Micro Devices (DMD) Lab, Department of Mechanical & Aerospace Engineering, Monash University, 17 College Walk, Clayton, VIC, 3168, Australia
| | - Takahide Murayama
- Institute of Semiconductor and Electronics Technologies ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Koukou Suu
- Institute of Semiconductor and Electronics Technologies ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Yasuhiro Morikawa
- Institute of Semiconductor and Electronics Technologies ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC, 3168, Australia
| | - Roey Elnathan
- School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, VIC, 3216, Australia
- Institute for Frontier Materials, Deakin University, Geelong Waurn Ponds campus, Waurn Ponds, VIC, 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong Waurn Ponds Campus, Melbourne, VIC, 3216, Australia
| |
Collapse
|
47
|
El-Sayed NNE, Al-Otaibi TM, Barakat A, Almarhoon ZM, Hassan MZ, Al-Zaben MI, Krayem N, Masand VH, Ben Bacha A. Synthesis and Biological Evaluation of Some New 3-Aryl-2-thioxo-2,3-dihydroquinazolin-4(1 H)-ones and 3-Aryl-2-(benzylthio)quinazolin-4(3 H)-ones as Antioxidants; COX-2, LDHA, α-Glucosidase and α-Amylase Inhibitors; and Anti-Colon Carcinoma and Apoptosis-Inducing Agents. Pharmaceuticals (Basel) 2023; 16:1392. [PMID: 37895863 PMCID: PMC10610505 DOI: 10.3390/ph16101392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Oxidative stress, COX-2, LDHA and hyperglycemia are interlinked contributing pathways in the etiology, progression and metastasis of colon cancer. Additionally, dysregulated apoptosis in cells with genetic alternations leads to their progression in malignant transformation. Therefore, quinazolinones 3a-3h and 5a-5h were synthesized and evaluated as antioxidants, enzymes inhibitors and cytotoxic agents against LoVo and HCT-116 cells. Moreover, the most active cytotoxic derivatives were evaluated as apoptosis inducers. The results indicated that 3a, 3g and 5a were efficiently scavenged DPPH radicals with lowered IC50 values (mM) ranging from 0.165 ± 0.0057 to 0.191 ± 0.0099, as compared to 0.245 ± 0.0257 by BHT. Derivatives 3h, 5a and 5h were recognized as more potent dual inhibitors than quercetin against α-amylase and α-glucosidase, in addition to 3a, 3c, 3f and 5b-5f against α-amylase. Although none of the compounds demonstrated a higher efficiency than the reference inhibitors against COX-2 and LDHA, 3a and 3g were identified as the most active derivatives. Molecular docking studies were used to elucidate the binding affinities and binding interactions between the inhibitors and their target proteins. Compounds 3a and 3f showed cytotoxic activities, with IC50 values (µM) of 294.32 ± 8.41 and 383.5 ± 8.99 (LoVo), as well as 298.05 ± 13.26 and 323.59 ± 3.00 (HCT-116). The cytotoxicity mechanism of 3a and 3f could be attributed to the modulation of apoptosis regulators (Bax and Bcl-2), the activation of intrinsic and extrinsic apoptosis pathways via the upregulation of initiator caspases-8 and -9 as well as executioner caspase-3, and the arrest of LoVo and HCT-116 cell cycles in the G2/M and G1 phases, respectively. Lastly, the physicochemical, medicinal chemistry and ADMET properties of all compounds were predicted.
Collapse
Affiliation(s)
| | - Taghreed M. Al-Otaibi
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (T.M.A.-O.); (A.B.); (M.I.A.-Z.)
| | - Assem Barakat
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (T.M.A.-O.); (A.B.); (M.I.A.-Z.)
| | - Zainab M. Almarhoon
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (T.M.A.-O.); (A.B.); (M.I.A.-Z.)
| | - Mohd. Zaheen Hassan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia;
| | - Maha I. Al-Zaben
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (T.M.A.-O.); (A.B.); (M.I.A.-Z.)
| | - Najeh Krayem
- Laboratoire de Biochimie et de Génie Enzymatique des Lipases, ENIS, Université de Sfax, Route de Soukra 3038, Sfax BP 1173, Tunisia;
| | - Vijay H. Masand
- Department of Chemistry, Vidya Bharati College, Camp, Amravati, Maharashtra 444602, India;
| | - Abir Ben Bacha
- Biochemistry Department, College of Sciences, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia;
| |
Collapse
|
48
|
Qian H, Baglamis S, Redeker F, Raaijman J, Hoebe RA, Sheraton VM, Vermeulen L, Krawczyk PM. High-Content and High-Throughput Clonogenic Survival Assay Using Fluorescence Barcoding. Cancers (Basel) 2023; 15:4772. [PMID: 37835466 PMCID: PMC10571559 DOI: 10.3390/cancers15194772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
The Clonogenic Survival Assay (CSA) is a fundamental tool employed to assess cell survival and proliferative potential in cancer research. Despite its importance, CSA faces limitations, primarily its time- and labor-intensive nature and its binary output. To overcome these limitations and enhance CSA's utility, several approaches have been developed, focusing on increasing the throughput. However, achieving both high-content and high-throughput analyses simultaneously has remained a challenge. In this paper, we introduce LeGO-CSA, an extension of the classical CSA that employs the imaging of cell nuclei barcoded with fluorescent lentiviral gene ontology markers, enabling both high-content and high-throughput analysis. To validate our approach, we contrasted it with results from a classical assay and conducted a proof-of-concept screen of small-molecule inhibitors targeting various pathways relevant to cancer treatment. Notably, our results indicate that the classical CSA may underestimate clonogenicity and unveil intriguing aspects of clonal cell growth. We demonstrate the potential of LeGO-CSA to offer a robust approach for assessing cell survival and proliferation with enhanced precision and throughput, with promising implications for accelerating drug discovery and contributing to a more comprehensive understanding of cellular behavior in cancer.
Collapse
Affiliation(s)
- Haibin Qian
- Department of Medical Biology, Amsterdam University Medical Centers (Location AMC), Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (H.Q.); (R.A.H.)
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
| | - Selami Baglamis
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, The Netherlands
| | - Fumei Redeker
- Department of Medical Biology, Amsterdam University Medical Centers (Location AMC), Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (H.Q.); (R.A.H.)
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
| | - Julia Raaijman
- Department of Medical Biology, Amsterdam University Medical Centers (Location AMC), Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (H.Q.); (R.A.H.)
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
| | - Ron A. Hoebe
- Department of Medical Biology, Amsterdam University Medical Centers (Location AMC), Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (H.Q.); (R.A.H.)
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
| | - Vivek M. Sheraton
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, The Netherlands
- Institute for Advanced Study, University of Amsterdam, 1012 WX Amsterdam, The Netherlands
| | - Louis Vermeulen
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, The Netherlands
| | - Przemek M. Krawczyk
- Department of Medical Biology, Amsterdam University Medical Centers (Location AMC), Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (H.Q.); (R.A.H.)
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
| |
Collapse
|
49
|
Li J, Han N, Li Y, Zhao F, Xiong W, Zeng Z. Evaluating the Antibacterial and Antivirulence Activities of Floxuridine against Streptococcus suis. Int J Mol Sci 2023; 24:14211. [PMID: 37762514 PMCID: PMC10532271 DOI: 10.3390/ijms241814211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Streptococcus suis is an emerging zoonotic pathogen that can cause fatal diseases such as meningitis and sepsis in pigs and human beings. The overuse of antibiotics is leading to an increased level of resistance in S. suis, and novel antimicrobial agents or anti-virulence agents for the treatment of infections caused by S. suis are urgently needed. In the present study, we investigated the antibacterial activity, mode of action and anti-virulence effects of floxuridine against S. suis. Floxuridine showed excessive antibacterial activity against S. suis both in vivo and in vitro; 4 × MIC of floxuridine could kill S. suis within 8 h in a time-kill assay. Meanwhile, floxuridine disrupted the membrane structure and permeability of the cytoplasmic membrane. Molecular docking revealed that floxuridine and SLY can be directly bind to each other. Moreover, floxuridine effectively inhibited the hemolytic capacity and expression levels of the virulence-related genes of S. suis. Collectively, these results indicate that the FDA-approved anticancer drug floxuridine is a promising agent and a potential virulence inhibitor against S. suis.
Collapse
Affiliation(s)
- Jie Li
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (W.X.)
- National Laboratory of Safety Evaluation (Environmental Assessment) of Veterinary Drugs, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Ning Han
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (W.X.)
- National Laboratory of Safety Evaluation (Environmental Assessment) of Veterinary Drugs, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yangyang Li
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (W.X.)
- National Laboratory of Safety Evaluation (Environmental Assessment) of Veterinary Drugs, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Feifei Zhao
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (W.X.)
- National Laboratory of Safety Evaluation (Environmental Assessment) of Veterinary Drugs, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Wenguang Xiong
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (W.X.)
- National Laboratory of Safety Evaluation (Environmental Assessment) of Veterinary Drugs, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhenling Zeng
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (W.X.)
- National Laboratory of Safety Evaluation (Environmental Assessment) of Veterinary Drugs, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
50
|
Osman NAHK, Abd-Elazeem OM, Al-Eisa RA, El-Shenawy NS. Anticancer and antimicrobial evaluation of extract from brown algae Hormophysa cuneiformis. J Appl Biomed 2023; 21:121-136. [PMID: 37747312 DOI: 10.32725/jab.2023.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023] Open
Abstract
AIM We investigated the antimicrobial and anticancer properties of an ethanol crude extract of Red Sea brown alga (Hormophysa cuneiformis) from Egypt. METHODS Extraction was achieved by mixing 100 g of sample powder with absolute ethanol, incubating at 37 °C overnight in a shaking incubator, and then collecting the extract. The extract's antimicrobial activity was tested using a well diffusion assay against the tested pathogens (Escherichia coli, Bacillus subtilis, Staphylococcus aureus, and Candida albicans) in comparison to commercial antibiotics. Anticancer activity was assessed using MTT assay on MCF-7, HepG-2, and HEP-2 cell lines. The anticancer mechanism of action against the HepG-2 cell line was investigated using cell cycle analysis, Annexin V, and antioxidant enzymes, in addition to transmission electron microscopy. RESULTS GC-MS phytoconstituent profile of the extract was dominant with fatty acids. A broad antimicrobial effect against all the pathogenic isolates of E. coli, S. aureus, B. subtitles, and C. albicans was demonstrated, especially at the high concentration in comparison to commercial antibiotics. The extract could inhibit the growth of the tested cell lines. We observed the most significant effect on HepG-2 cells, and the concentration of the extract played a role in the level of inhibition (IC50 of 44.6 ± 0.6 µg/ml). The extract had negligible effects on Vero normal cell lines at the lower concentration, with slight toxicity (90.8% viability) at the highest concentration (500 µg/ml). At this same concentration, the extract caused 80-92% inhibition of the cancer cell lines. The extract appears to have demonstrated promising effects on cancer cells. It induces programmed cell death (apoptosis), arrests the cell cycle, and affects the oxidative/antioxidant balance within the cells, potentially leading to the suppression or elimination of cancer cells. These findings are encouraging and may have implications for cancer treatment or further research in this area. More action of extract was seen against bacteria than fungi, with a wide antibacterial impact against all of the tested isolates, notably at the high concentration in comparison to conventional antibiotics. CONCLUSION According to the findings, H. cuneiformis may be a valuable source of chemicals that are both antimicrobial and anticancer.
Collapse
Affiliation(s)
- Nehal A H K Osman
- Suez Canal University, Faculty of Science, Botany and Microbiology Department, Ismailia 41522, Egypt
| | - Omniya M Abd-Elazeem
- Suez Canal University, Faculty of Science, Department of Zoology, Ismailia 41522, Egypt
| | - Rasha A Al-Eisa
- Taif University, College of Sciences, Department of Biology, Taif 21944, Saudi Arabia
| | - Nahla S El-Shenawy
- Suez Canal University, Faculty of Science, Department of Zoology, Ismailia 41522, Egypt
| |
Collapse
|