1
|
Roy S, Paul S, Mukherjee S, De P, Mukherjee A. Unraveling Mechanism and Enhancing Selectivity of a Ru II-bis-bipyridyl-morphocumin Complex with RAFT-Generated Glycopolymer Exploiting Warburg Effect in Cancer. Chemistry 2024:e202403695. [PMID: 39614769 DOI: 10.1002/chem.202403695] [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: 10/26/2024] [Indexed: 12/12/2024]
Abstract
The Warburg effect, which generates increased demand of glucose in cancer cells is a relatively underexplored phenomenon in existing commercial drugs to enhance uptake in cancer cells. Here, we present a chemotherapeutic strategy employing a Ru(II)-bis-bipyridyl-morphocumin complex (2) encapsulated in a self-assembling glucose-functionalized copolymer P(G-EMA-co-MMA) (where G=glucose; MMA=methyl methacrylate; EMA=ethyl methacrylate), designed to exploit this effect for enhanced selectivity in cancer treatment. The P(G-EMA-co-MMA) polymer, synthesized via reversible-addition fragmentation chain transfer (RAFT) polymerization, has a number average molecular weight (Mn,NMR) of 8000 g/mol. Complex 2, stable in aqueous media, selectively releases a cytotoxic, lysosome-targeting compound, morphocumin, in the presence of excess hydrogen peroxide (H₂O₂), a reactive oxygen species (ROS) prevalent in tumor microenvironments. Additionally, complex 2 promotes ROS accumulation, which may further enhance morphocumin release through a synergistic domino effect. Comparative studies reveal that 2 outperforms its curcumin Ru(II) complex (1) analog in solution stability, organelle specificity, and cellular mechanisms. Both 1 and 2 exhibit phototherapeutic effects under low-intensity visible light, but their chemotoxicity significantly increases with incubation time in the dark, highlighting the superior chemotherapeutic efficacy of the O,O-coordinating Ru(II) ternary polypyridyl complexes. Complex 2 induces apoptosis via the intrinsic pathway and shows a 9-fold increase in selectivity for pancreatic cancer cells (MIA PaCa-2) over non-cancerous HEK293 cells when encapsulated in the glucose-conjugated polymer (DP@2). Glucose deprivation in the culture medium further enhances drug efficacy by an additional 5-fold. This work underscores the potential of glucose-functionalized polymers and ROS-responsive Ru(II) complexes in targeted cancer therapy.
Collapse
Affiliation(s)
- Souryadip Roy
- Centre for Advanced Functional Materials, Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Nadia, West Bengal, Mohanpur, 741246, India
| | - Soumya Paul
- Centre for Advanced Functional Materials, Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Nadia, West Bengal, Mohanpur, 741246, India
| | - Sujato Mukherjee
- Centre for Advanced Functional Materials, Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Nadia, West Bengal, Mohanpur, 741246, India
| | - Priyadarsi De
- Centre for Advanced Functional Materials, Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Nadia, West Bengal, Mohanpur, 741246, India
| | - Arindam Mukherjee
- Centre for Advanced Functional Materials, Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Nadia, West Bengal, Mohanpur, 741246, India
| |
Collapse
|
2
|
Mukherjee D, Raikwar S. Recent Update on Nanocarrier(s) as the Targeted Therapy for Breast Cancer. AAPS PharmSciTech 2024; 25:153. [PMID: 38961013 DOI: 10.1208/s12249-024-02867-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 06/11/2024] [Indexed: 07/05/2024] Open
Abstract
Despite ongoing advances in cancer therapy, the results for the treatment of breast cancer are not satisfactory. The advent of nanotechnology promises to be an essential tool to improve drug delivery effectiveness in cancer therapy. Nanotechnology provides an opportunity to enhance the treatment modality by preventing degradation, improving tumour targeting, and controlling drug release. Recent advances have revealed several strategies to prevent cancer metastasis using nano-drug delivery systems (NDDS). These strategies include the design of appropriate nanocarriers loaded with anti-cancer drugs that target the optimization of physicochemical properties, modulate the tumour microenvironment, and target biomimetic techniques. Nanocarriers have emerged as a preferential approach in the chemotropic treatment for breast cancer due to their pivotal role in safeguarding the therapeutic agents against degradation. They facilitate efficient drug concentration in targeted cells, surmount the resistance of drugs, and possess a small size. Nevertheless, these nanocarrier(s) have some limitations, such as less permeability across the barrier and low bioavailability of loaded drugs. To overcome these challenges, integrating external stimuli has been employed, encompassing infrared light, thermal stimulation, microwaves, and X-rays. Among these stimuli, ultrasound-triggered nanocarriers have gained significant attention due to their cost-effectiveness, non-invasive nature, specificity, ability to penetrate tissues, and capacity to deliver elevated drug concentrations to intended targets. This article comprehensively reviews recent advancements in different nanocarriers for breast cancer chemotherapy. It also delves into the associated hurdles and offers valuable insights into the prospective directions for this innovative field.
Collapse
Affiliation(s)
- Debanjan Mukherjee
- Department of Quality Assurance, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Sarjana Raikwar
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
3
|
Berckmans Y, Ene HM, Ben-Meir K, Martinez-Conde A, Wouters R, Van den Ende B, Van Mechelen S, Monin R, Frechtel-Gerzi R, Gabay H, Dor-On E, Haber A, Weinberg U, Vergote I, Giladi M, Coosemans A, Palti Y. Tumor Treating Fields (TTFields) induce homologous recombination deficiency in ovarian cancer cells, thus mitigating drug resistance. Front Oncol 2024; 14:1402851. [PMID: 38993641 PMCID: PMC11238040 DOI: 10.3389/fonc.2024.1402851] [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: 03/18/2024] [Accepted: 06/10/2024] [Indexed: 07/13/2024] Open
Abstract
Background Ovarian cancer is the leading cause of mortality among gynecological malignancies. Carboplatin and poly (ADP-ribose) polymerase inhibitors (PARPi) are often implemented in the treatment of ovarian cancer. Homologous recombination deficient (HRD) tumors demonstrate increased sensitivity to these treatments; however, many ovarian cancer patients are homologous recombination proficient (HRP). TTFields are non-invasive electric fields that induce an HRD-like phenotype in various cancer types. The current study aimed to investigate the impact of TTFields applied together with carboplatin or PARPi (olaparib or niraparib) in preclinical ovarian cancer models. Methods A2780 (HRP), OVCAR3 (HRD), and A2780cis (platinum-resistant) human ovarian cancer cells were treated in vitro with TTFields (1 V/cm RMS, 200 kHz, 72 h), alone or with various drug concentrations. Treated cells were measured for cell count, colony formation, apoptosis, DNA damage, expression of DNA repair proteins, and cell cycle. In vivo, ID8-fLuc (HRP) ovarian cancer cells were inoculated intraperitoneally to C57BL/6 mice, which were then treated with either sham, TTFields (200 kHz), olaparib (50 mg/kg), or TTFields plus olaparib; over a period of four weeks. Tumor growth was analyzed using bioluminescent imaging at treatment cessation; and survival analysis was performed. Results The nature of TTFields-drug interaction was dependent on the drug's underlying mechanism of action and on the genetic background of the cells, with synergistic interactions between TTFields and carboplatin or PARPi seen in HRP and resistant cells. Treated cells demonstrated elevated levels of DNA damage, accompanied by G2/M arrest, and induction of an HRD-like phenotype. In the tumor-bearing mice, TTFields and olaparib co-treatment resulted in reduced tumor volume and a survival benefit relative to olaparib monotherapy and to control. Conclusion By inducing an HRD-like phenotype, TTFields sensitize HRP and resistant ovarian cancer cells to treatment with carboplatin or PARPi, potentially mitigating a-priori and de novo drug resistance, a major limitation in ovarian cancer treatment.
Collapse
Affiliation(s)
- Yani Berckmans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | | | | | | | - Roxanne Wouters
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
- Oncoinvent AS, Oslo, Norway
| | - Bieke Van den Ende
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Sara Van Mechelen
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | | - Ignace Vergote
- Department of Gynecology and Obstetrics, Gynecologic Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | | | - An Coosemans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
4
|
Goldstien L, Lavi Y, Atia L. ConfluentFUCCI for fully-automated analysis of cell-cycle progression in a highly dense collective of migrating cells. PLoS One 2024; 19:e0305491. [PMID: 38924026 PMCID: PMC11207131 DOI: 10.1371/journal.pone.0305491] [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: 11/07/2023] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Understanding mechanisms underlying various physiological and pathological processes often requires accurate and fully automated analysis of dense cell populations that collectively migrate. In such multicellular systems, there is a rising interest in the relations between biophysical and cell cycle progression aspects. A seminal tool that led to a leap in real-time study of cell cycle is the fluorescent ubiquitination-based cell cycle indicator (FUCCI). Here, we introduce ConfluentFUCCI, an open-source graphical user interface-based framework that is designed, unlike previous tools, for fully automated analysis of cell cycle progression, cellular dynamics, and cellular morphology, in highly dense migrating cell collectives. We integrated into ConfluentFUCCI's pipeline state-of-the-art tools such as Cellpose, TrackMate, and Napari, some of which incorporate deep learning, and we wrap the entire tool into an isolated computational environment termed container. This provides an easy installation and workflow that is independent of any specific operation system. ConfluentFUCCI offers accurate nuclear segmentation and tracking using FUCCI tags, enabling comprehensive investigation of cell cycle progression at both the tissue and single-cell levels. We compare ConfluentFUCCI to the most recent relevant tool, showcasing its accuracy and efficiency in handling large datasets. Furthermore, we demonstrate the ability of ConfluentFUCCI to monitor cell cycle transitions, dynamics, and morphology within densely packed epithelial cell populations, enabling insights into mechanotransductive regulation of cell cycle progression. The presented tool provides a robust approach for investigating cell cycle-related phenomena in complex biological systems, offering potential applications in cancer research and other fields.
Collapse
Affiliation(s)
- Leo Goldstien
- Department of Mechanical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yael Lavi
- Department of Mechanical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Lior Atia
- Department of Mechanical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
5
|
Cen JH, Xie QH, Guo GH, Xu SY, Liu ZY, Liao YH, Zhong XP, Liu HY. Construction of 5-Fluorouracil and Gallium Corrole Conjugates for Enhanced Photodynamic Therapy. J Med Chem 2024; 67:9054-9068. [PMID: 38781403 DOI: 10.1021/acs.jmedchem.4c00249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Molecular hybridization is a well-established strategy for developing new drugs. In the pursuit of promising photosensitizers (PSs) with enhanced photodynamic therapy (PDT) efficiency, a series of novel 5-fluorouracil (5FU) gallium corrole conjugates (1-Ga-4-Ga) were designed and synthesized by hybridizing a chemotherapeutic drug and PSs. Their photodynamic antitumor activity was also evaluated. The most active complex (2-Ga) possesses a low IC50 value of 0.185 μM and a phototoxic index of 541 against HepG2 cells. Additionally, the 5FU-gallium corrole conjugate (2-Ga) exhibited a synergistic increase in cytotoxicity under irradiation. Excitedly, treatment of HepG2 tumor-bearing mice with 2-Ga under irradiation could completely ablate tumors without harming normal tissues. 2-Ga-mediated PDT could disrupt mitochondrial function, cause cell cycle arrest in the sub-G1 phase, and activate the cell apoptosis pathway by upregulating the cleaved PARP expression and the Bax/Bcl-2 ratios. This work provides a useful strategy for the design of new corrole-based chemo-photodynamic therapy drugs.
Collapse
Affiliation(s)
- Jing-He Cen
- School of Chemistry and Chemical Engineering, The Key Laboratory of Fuel Cell Technology of Guangdong Province, South China University of Technology, Guangzhou 510641, China
| | - Qi-Hu Xie
- Department of Plastic Surgery and Burns, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Geng-Hong Guo
- Department of Plastic Surgery and Burns, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Shi-Yin Xu
- School of Chemistry and Chemical Engineering, The Key Laboratory of Fuel Cell Technology of Guangdong Province, South China University of Technology, Guangzhou 510641, China
| | - Ze-Yu Liu
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
| | - Yu-Hui Liao
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
| | - Xiao-Ping Zhong
- Department of Plastic Surgery and Burns, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Hai-Yang Liu
- School of Chemistry and Chemical Engineering, The Key Laboratory of Fuel Cell Technology of Guangdong Province, South China University of Technology, Guangzhou 510641, China
| |
Collapse
|
6
|
Joshi AS, Bapat MV, Singh P, Mijakovic I. Viridibacillus culture derived silver nanoparticles exert potent anticancer action in 2D and 3D models of lung cancer via mitochondrial depolarization-mediated apoptosis. Mater Today Bio 2024; 25:100997. [PMID: 38379934 PMCID: PMC10876681 DOI: 10.1016/j.mtbio.2024.100997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/03/2024] [Accepted: 02/09/2024] [Indexed: 02/22/2024] Open
Abstract
Lung cancer is one of the most commonly occurring cancer types that accounts for almost 2 million cases per year. Its resistance to anticancer drugs, failure of new molecules in clinical trials, severe side-effects of current treatments, and its recurrence limit the success of anticancer therapies. Nanotherapeutic agents offer several advantages over conventional anticancer therapies, including improved retention in tumors, specificity, and anticancer effects at lower concentrations, hence reducing the side-effects. Here, we have explored the anticancer activity of silver nanoparticles synthesized in Viridibacillus sp. enriched culture medium for the first time. Such green nanoparticles, synthesized by biological systems, are superior to chemically synthesized ones in terms of their environmental footprint and production cost, and have one crucial advantage of excellent stability owing to their biological corona. To assess anticancer activity of these nanoparticles, we used conventional 2D cultured A549 cells as well as 3D spheroids of A549 cells. In both models of lung cancer, our silver nanoparticles diminished cell proliferation, arrested DNA synthesis, and showed a dose dependent cytotoxic effect. The nanoparticles damaged the DNA and mitochondrial structures in both A549 cells and A549 spheroids, leading to mitochondrial depolarization and increased cell permeability. Low lethal median doses (LD50) for 2D cultured A549 cells (1 μg/ml) and for A549 spheroids (13 μg/ml) suggest that our nanoparticles are potent anticancer agents. We also developed in vitro tumor progression model and in vitro tumor size model using 3D spheroids to test anticancer potential of our nanoparticles which otherwise would require longer experimental duration along with large number of animals and trained personnel. In these models, our nanoparticles showed strong dose dependent anticancer activity. In case of in vitro tumor progression model, the A549 cells failed to form tight spheroidal mass and showed increased dead cell fraction since day 1 as compared to control. On the other hand, in case of in vitro tumor size model, the 4 and 8 μg/ml nanoparticle treatment led to reduction in spheroid size from 615 ± 53 μm to 440 ± 45 μm and 612 ± 44 μm to 368 ± 62 μm respectively, within the time span of 3 days post treatment. We believe that use of such novel experimental models offers excellent and fast alternative to in vivo studies, and to the best of our knowledge, this is the first report that gives proof-of-concept for use of such novel in vitro cancer models to test anticancer agents such as Viridibacilli culture derived silver nanoparticles. Based on our results, we propose that these nanoparticles offer an interesting alternative for anticancer therapies, especially if they can be combined with classical anticancer drugs.
Collapse
Affiliation(s)
- Abhayraj S. Joshi
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Mugdha V. Bapat
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Priyanka Singh
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Ivan Mijakovic
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
- Department of Biology and Biological Engineering, Division of Systems and Synthetic Biology, Chalmers University of Technology, Sweden
| |
Collapse
|
7
|
Mohamed Azar KAH, Ezhilarasan D, Shree Harini K. Coleus vettiveroides ethanolic root extract induces cytotoxicity by intrinsic apoptosis in HepG2 cells. J Appl Toxicol 2024; 44:245-259. [PMID: 37661188 DOI: 10.1002/jat.4536] [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: 04/19/2023] [Revised: 08/08/2023] [Accepted: 08/14/2023] [Indexed: 09/05/2023]
Abstract
Hepatocellular carcinoma (HCC) contributes to more than 80% of all primary cancers globally and ranks fourth in cancer-related deaths, due to the lack of an effective, definite therapeutic drug. Coleus vettiveroides (CV) has been used in Indian traditional medicine to treat diabetes, liver ailments, skin diseases, leukoderma, and leprosy. This study investigates the anticancer effect of CV ethanolic root extract in HepG2 cells. HepG2 cells were treated with CV extract, and its cytotoxicity was analyzed by MTT assay. AO/EB staining, propidium iodide staining, DCFH-DA assay, phalloidine staining, flow cytometry, and qPCR studies were performed for ROS expression, apoptosis and cell cycle analysis. The phytochemical analysis confirmed the presence of quercetin and galangin in CV root extract. The results showed that CV inhibited the proliferation of HepG2 cells, with altered cellular and nuclear morphology. CV was also found to increase intracellular ROS levels and oxidative stress markers in HepG2 cells. CV significantly altered the actin microfilament distribution in HepG2 cells and caused cell cycle arrest at the sub G0 -G1 phase. CV also induced mitochondria-mediated apoptosis, as evidenced by increased expression of p53, Bax, cytochrome C, Apaf-1, PARP, caspase-3 and caspase-9, and downregulated Bcl-2 expression. Therefore, CV exerts its anticancer effect by inducing mitochondrial dysfunction, oxidative stress, cytoskeletal disorganization, cell cycle arrest, and mitochondria-mediated apoptosis, and it could be a potent therapeutic option for HCC.
Collapse
Affiliation(s)
- Kadmad Abdul Hameed Mohamed Azar
- Department of Pharmacology, Koppal Institute of Medical Sciences, Koppal, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| | - Devaraj Ezhilarasan
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| | - Karthik Shree Harini
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| |
Collapse
|
8
|
Sharlow ER, Llaneza DC, Tewari BP, Mingledorff GA, Mendelson AJ, Sontheimer H, Bloom GS, Lazo JS. Pharmacological profiling identifies divergent chemosensitivities of differentiating and maturing iPSC-derived human cortical neuron populations. FEBS J 2023; 290:4950-4965. [PMID: 37428551 PMCID: PMC10592385 DOI: 10.1111/febs.16901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 06/16/2023] [Accepted: 07/07/2023] [Indexed: 07/11/2023]
Abstract
Neuronal differentiation and maturation are extended developmental processes. To determine whether neurons at different developmental stages have divergent chemosensitivities, we screened differentiating and maturing neuronal populations using a small compound library comprising FDA-approved and investigational drugs. Using a neurotoxicity assay format, both respective neuronal population-based screening campaigns performed robustly (Z-factors = 0.7-0.8), although the hit rate for the differentiating neurons (2.8%) was slightly higher than for maturing neurons (1.9%). While the majority of hits were toxic to both neuronal populations, these hits predominantly represented promiscuous drugs. Other drugs were selectively neurotoxic, with receptor tyrosine kinase inhibitors disproportionally represented after confirmation. Ponatinib and amuvatinib were neuroinhibitory for differentiating and maturing neurons, respectively. Chemoinformatic analyses confirmed differences in potential drug targets that may be differentially expressed during neuronal development. Subsequent studies demonstrated neuronal expression of AXL, an amuvatinib target, in both neuronal populations. However, functional AXL activity was confirmed only in the maturing neuronal population as determined by AXL phosphorylation in response to GAS6, the cognate ligand of AXL, and concurrent STAT3Y705 phosphorylation. Differentiating neurons were unresponsive to the effects of GAS6 suggesting that the AXL-STAT3 signaling axis was nonfunctional. Amuvatinib treatment of maturing neuronal cultures significantly reduced pAXL levels. These studies indicate that neuronal developmental states may exhibit unique chemosensitivities and that drugs may have different neuro-inhibitory effects depending upon the developmental stage of the neuronal population.
Collapse
Affiliation(s)
| | - Danielle C. Llaneza
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908
| | - Bhanu P. Tewari
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22908
| | | | - Anna J. Mendelson
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908
| | - Harald Sontheimer
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22908
| | - George S. Bloom
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22908
- Department of Biology, University of Virginia, Charlottesville, VA 22904
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908
| | - John S. Lazo
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908
| |
Collapse
|
9
|
Menghini S, Vizovisek M, Enders J, Schuerle S. Magnetospirillum magneticum triggers apoptotic pathways in human breast cancer cells. Cancer Metab 2023; 11:12. [PMID: 37559137 PMCID: PMC10410830 DOI: 10.1186/s40170-023-00313-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 07/24/2023] [Indexed: 08/11/2023] Open
Abstract
The use of bacteria in cancer immunotherapy has the potential to bypass many shortcomings of conventional treatments. The ability of anaerobic bacteria to preferentially accumulate and replicate in hypoxic regions of solid tumors, as a consequence of bacterial metabolic needs, is particularly advantageous and key to boosting their immunostimulatory therapeutic actions in situ. While several of these bacterial traits are well-studied, little is known about their competition for nutrients and its effect on cancer cells which could serve as another potent and innate antineoplastic action. Here, we explored the consequences of the iron-scavenging abilities of a particular species of bacteria, Magnetospirillum magneticum, which has been studied as a potential new class of bacteria for magnetically targeted bacterial cancer therapy. We investigated their influence in hypoxic regions of solid tumors by studying the consequential metabolic effects exerted on cancer cells. To do so, we established an in vitro co-culture system consisting of the bacterial strain AMB-1 incubated under hypoxic conditions with human breast cancer cells MDA-MB-231. We first quantified the number of viable cells after incubation with magnetotactic bacteria demonstrating a lower rate of cellular proliferation that correlated with increasing bacteria-to-cancer cells ratio. Further experiments showed increasing populations of apoptotic cells when cancer cells were incubated with AMB-1 over a period of 24 h. Analysis of the metabolic effects induced by bacteria suggest an increase in the activation of executioner caspases as well as changes in levels of apoptosis-related proteins. Finally, the level of several human apoptosis-related proteins was investigated, confirming a bacteria-dependent triggering of apoptotic pathways in breast cancer cells. Overall, our findings support that magnetotactic bacteria could act as self-replicating iron-chelating agents and indicate that they interfere with proliferation and lead to increased apoptosis of cancer cells. This bacterial feature could serve as an additional antineoplastic mechanism to reinforce current bacterial cancer therapies.
Collapse
Affiliation(s)
- Stefano Menghini
- Department of Health Sciences and Technology, Institute for Translational Medicine, ETH Zurich, CH-8092, Zurich, Switzerland
| | - Matej Vizovisek
- Department of Health Sciences and Technology, Institute for Translational Medicine, ETH Zurich, CH-8092, Zurich, Switzerland
| | - Jonathas Enders
- Department of Health Sciences and Technology, Institute for Translational Medicine, ETH Zurich, CH-8092, Zurich, Switzerland
| | - Simone Schuerle
- Department of Health Sciences and Technology, Institute for Translational Medicine, ETH Zurich, CH-8092, Zurich, Switzerland.
| |
Collapse
|
10
|
Mohammadi A, Mansoori B, Safarzadeh E, Gholizadeh S, Baradaran B. Anacyclus pyrethrum extract significantly destroyed lung cancer cell line (A549) by inducing apoptosis. J Herb Med 2023. [DOI: 10.1016/j.hermed.2023.100649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
11
|
Kim H, Hwang B, Cho S, Kim WJ, Myung SC, Choi YH, Kim WJ, Lee S, Moon SK. The ethanol extract of Cyperus exaltatus var. iwasakii exhibits cell cycle dysregulation, ERK1/2/p38 MAPK/AKT phosphorylation, and reduced MMP-9-mediated metastatic capacity in prostate cancer models in vitro and in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154794. [PMID: 37011419 DOI: 10.1016/j.phymed.2023.154794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Prostate cancer is the second most common cause of cancer death worldwide in men. The development of novel and highly efficient therapeutic strategies is strongly recommended to treat prostate cancer. Cyperaceae are an ecologically and economically important family of plants with several pharmacological effects. However, the biological efficacy of Cyperus exaltatus var. iwasakii (CE) is unknown. PURPOSE This study aimed to investigate the antitumor effect of the ethanol extract of CE against prostate cancer. METHODS In vitro antitumor efficacy of CE was explored by the MTT assay, cell counting assay, FACS analysis, immunoblot, wound-healing migration, invasion assay, zymographic assay, and EMSA in prostate cancer cells, DU145 and LNCaP. For in vivo experiments, xenograft mice were injected with LNCaP cells. Histology (H&E and Ki-67) and biochemical enzyme assay were then performed. The toxicity test was evaluated by an acute toxicity assay. The phytochemical constituents of CE were identified by spectrometric and chromatographic analyses. RESULTS CE exerted a significant antiproliferative effect against prostate cancer cells. CE-induced antiproliferative cells were associated with cell cycle arrest at G0/G1 (cyclin D1/CDK4, cyclin E/CDK2, p21Waf1) in DU145 cells, but G2/M (ATR, CHK1, Cdc2, Cdc25c, p21Waf1, and p53) in LNCaP cells. CE stimulated the phosphorylation of ERK1/2, p38 MAPK, and AKT in DU145 cells, but only p38 MAPK phosphorylation was increased in LNCaP cells. CE treatment suppressed migration and invasion in the two types of prostate cancer cells by inhibiting MMP-9 activity through the regulation of transcription factors, such as AP-1 and NF-κB. In vivo experiments showed a reduction in tumor weight and size following oral CE administration. Histochemistry confirmed that CE inhibited tumor growth in the mouse LNCaP xenograft model. The administration of CE had no adverse effects on body weight, behavioral patterns, blood biochemistry, and histopathology findings of vital organs in mice. Finally, a total of 13 phytochemical constituents were identified and quantified in CE. The most abundant secondary metabolites in CE were astragalin, tricin, and p-coumaric acid. CONCLUSION Our results demonstrated the antitumor efficacy of CE against prostate cancer. These findings suggest that CE might be a potential candidate for prostate cancer prevention or treatment.
Collapse
Affiliation(s)
- Hoon Kim
- Department of Food and Nutrition, Chung-Ang University, 4726 Seodong-Daero, Daedeok-Myeon, Anseong 17546, Republic of Korea
| | - Byungdoo Hwang
- Department of Food and Nutrition, Chung-Ang University, 4726 Seodong-Daero, Daedeok-Myeon, Anseong 17546, Republic of Korea
| | - Seongbin Cho
- Department of Food and Nutrition, Chung-Ang University, 4726 Seodong-Daero, Daedeok-Myeon, Anseong 17546, Republic of Korea
| | - Woo Jung Kim
- Biocenter, Gyeonggido Business and Science Accelerator, Suwon 16229, Republic of Korea
| | - Soon Chul Myung
- Department of Urology, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dongeui University, Busan 47340, Republic of Korea
| | - Wun-Jae Kim
- Institute of Urotech, Cheongju, Chungcheongbuk-do 361-763, Republic of Korea
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, 4726 Seodong-Daero, Daedeok-Myeon, Anseong 17546, Republic of Korea.
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung-Ang University, 4726 Seodong-Daero, Daedeok-Myeon, Anseong 17546, Republic of Korea.
| |
Collapse
|
12
|
Mughal MJ, Bhadresha K, Kwok HF. CDK inhibitors from past to present: A new wave of cancer therapy. Semin Cancer Biol 2023; 88:106-122. [PMID: 36565895 DOI: 10.1016/j.semcancer.2022.12.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Deregulation of the cell cycle machinery, which has been linked to dysregulation of cyclin-dependent kinases (CDKs), is a defining characteristic of cancer, eventually promoting abnormal proliferation that feeds tumorigenesis and disease development. In this regard, several CDK inhibitors (CDKIs) have been developed during the last few decades (1st, 2nd, and 3rd generation CDKIs) to inhibit cancer cell proliferation. 1st and 2nd generation CDKIs have not received much clinical attention for the treatment of cancer patients because of their limited specificity and high toxicity. However, the recent development of combination strategies allowed us to reduce the toxicity and side effects of these CDKIs, paving the way for their potential application in clinical settings. The 3rd generation CDKIs have yielded the most promising results at the preclinical and clinical levels, propelling them into the advanced stages of clinical trials against multiple malignancies, especially breast cancer, and revolutionizing traditional treatment strategies. In this review, we discuss the most-investigated candidates from the 1st, 2nd, and 3rd generations of CDKIs, their basic mechanisms of action, the reasons for their failure in the past, and their current clinical development for the treatment of different malignancies. Additionally, we briefly highlighted the most recent clinical trial results and advances in the development of 3rd generation FDA-approved selective CDK4/6 inhibitors that combat the most prevalent cancer. Overall, this review will provide a thorough knowledge of CDKIs from the past to the present, allowing researchers to rethink and develop innovative cancer therapeutic regimens.
Collapse
Affiliation(s)
- Muhammad Jameel Mughal
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; MOE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC, United States
| | - Kinjal Bhadresha
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; Hematology/Oncology Division, School of Medicine, Indiana University Indianapolis, IN, United States
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; MOE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR.
| |
Collapse
|
13
|
Wang Z, Ouyang G, Liu W, Hou M, Wang Q, Tan H, Ji L, Wang C, Long X. Design, Synthesis and Biological Evaluation of Pyrimidinamine Derivatives Containing Urea Moiety. HETEROCYCLES 2023. [DOI: 10.3987/com-22-14745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
14
|
Lu Y, Massicano AVF, Gallegos CA, Heinzman KA, Parish SW, Warram JM, Sorace AG. Evaluating the Accuracy of FUCCI Cell Cycle In Vivo Fluorescent Imaging to Assess Tumor Proliferation in Preclinical Oncology Models. Mol Imaging Biol 2022; 24:898-908. [PMID: 35650411 DOI: 10.1007/s11307-022-01739-9] [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: 11/16/2021] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 12/29/2022]
Abstract
PURPOSE The primary goal of this study is to evaluate the accuracy of the fluorescence ubiquitination cell cycle indicator (FUCCI) system with fluorescence in vivo imaging compared to 3'-deoxy-3'-[18F]fluorothymidine ([18F]-FLT) positron emission tomography (PET)/computed tomography (CT) and biological validation through histology. Imaging with [18F]-FLT PET/CT can be used to noninvasively assess cancer cell proliferation and has been utilized in both preclinical and clinical studies. However, a cost-effective and straightforward method for in vivo, cell cycle targeted cancer drug screening is needed prior to moving towards translational imaging methods such as PET/CT. PROCEDURES In this study, fluorescent MDA-MB-231-FUCCI tumor growth was monitored weekly with caliper measurements and fluorescent imaging. Seven weeks post-injection, [18F]-FLT PET/CT was performed with a preclinical PET/CT, and tumors samples were harvested for histological analysis. RESULTS RFP fluorescent signal significantly correlated with tumor volume (r = 0.8153, p < 0.0001). Cell proliferation measured by GFP fluorescent imaging was correlated with tumor growth rate (r = 0.6497, p < 0.001). Also, GFP+ cells and [18F]-FLT regions of high uptake were both spatially located in the tumor borders, indicating that the FUCCI-IVIS method may provide an accurate assessment of tumor heterogeneity of cell proliferation. The quantification of total GFP signal was correlated with the sum of tumor [18F]-FLT standard uptake value (SUV) (r = 0.5361, p = 0.0724). Finally, histological analysis confirmed viable cells in the tumor and the correlation of GFP + and Ki67 + cells (r = 0.6368, p = 0.0477). CONCLUSION Fluorescent imaging of the cell cycle provides a noninvasive accurate depiction of tumor progression and response to therapy, which may benefit in vivo testing of novel cancer therapeutics that target the cell cycle.
Collapse
Affiliation(s)
- Yun Lu
- Department of Radiology, University of Alabama at Birmingham, Volker Hall G082, 1670 University Boulevard, Birmingham, AL, 35233, USA
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Adriana V F Massicano
- Department of Radiology, University of Alabama at Birmingham, Volker Hall G082, 1670 University Boulevard, Birmingham, AL, 35233, USA
| | - Carlos A Gallegos
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Katherine A Heinzman
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Sean W Parish
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Jason M Warram
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Anna G Sorace
- Department of Radiology, University of Alabama at Birmingham, Volker Hall G082, 1670 University Boulevard, Birmingham, AL, 35233, USA.
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|
15
|
Giannareas N, Zhang Q, Yang X, Na R, Tian Y, Yang Y, Ruan X, Huang D, Yang X, Wang C, Zhang P, Manninen A, Wang L, Wei GH. Extensive germline-somatic interplay contributes to prostate cancer progression through HNF1B co-option of TMPRSS2-ERG. Nat Commun 2022; 13:7320. [PMID: 36443337 PMCID: PMC9705428 DOI: 10.1038/s41467-022-34994-z] [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: 06/18/2021] [Accepted: 11/15/2022] [Indexed: 11/29/2022] Open
Abstract
Genome-wide association studies have identified 270 loci conferring risk for prostate cancer (PCa), yet the underlying biology and clinical impact remain to be investigated. Here we observe an enrichment of transcription factor genes including HNF1B within PCa risk-associated regions. While focused on the 17q12/HNF1B locus, we find a strong eQTL for HNF1B and multiple potential causal variants involved in the regulation of HNF1B expression in PCa. An unbiased genome-wide co-expression analysis reveals PCa-specific somatic TMPRSS2-ERG fusion as a transcriptional mediator of this locus and the HNF1B eQTL signal is ERG fusion status dependent. We investigate the role of HNF1B and find its involvement in several pathways related to cell cycle progression and PCa severity. Furthermore, HNF1B interacts with TMPRSS2-ERG to co-occupy large proportion of genomic regions with a remarkable enrichment of additional PCa risk alleles. We finally show that HNF1B co-opts ERG fusion to mediate mechanistic and biological effects of the PCa risk-associated locus 17p13.3/VPS53/FAM57A/GEMIN4. Taken together, we report an extensive germline-somatic interaction between TMPRSS2-ERG fusion and genetic variations underpinning PCa risk association and progression.
Collapse
Affiliation(s)
- Nikolaos Giannareas
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Qin Zhang
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Xiayun Yang
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Rong Na
- Division of Urology, Department of Surgery, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, China
| | - Yijun Tian
- Department of Tumour Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Yuehong Yang
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Xiaohao Ruan
- Department of Urology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Da Huang
- Department of Urology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaoqun Yang
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chaofu Wang
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Peng Zhang
- Fudan University Shanghai Cancer Center & MOE Key Laboratory of Metabolism and Molecular Medicine and Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Aki Manninen
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Liang Wang
- Department of Tumour Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Gong-Hong Wei
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland.
- Fudan University Shanghai Cancer Center & MOE Key Laboratory of Metabolism and Molecular Medicine and Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China.
| |
Collapse
|
16
|
Chibaya L, Snyder J, Ruscetti M. Senescence and the tumor-immune landscape: Implications for cancer immunotherapy. Semin Cancer Biol 2022; 86:827-845. [PMID: 35143990 PMCID: PMC9357237 DOI: 10.1016/j.semcancer.2022.02.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/03/2022] [Indexed: 01/27/2023]
Abstract
Cancer therapies, including conventional chemotherapy, radiation, and molecularly targeted agents, can lead to tumor eradication through a variety of mechanisms. In addition to their effects on tumor cell growth and survival, these regimens can also influence the surrounding tumor-immune microenvironment in ways that ultimately impact therapy responses. A unique biological outcome of cancer therapy is induction of cellular senescence. Senescence is a damage-induced stress program that leads to both the durable arrest of tumor cells and remodeling the tumor-immune microenvironment through activation of a collection pleiotropic cytokines, chemokines, growth factors, and proteinases known as the senescence-associated secretory phenotype (SASP). Depending on the cancer context and the mechanism of action of the therapy, the SASP produced following therapy-induced senescence (TIS) can promote anti-tumor immunity that enhances therapeutic efficacy, or alternatively chronic inflammation that leads to therapy failure and tumor relapse. Thus, a deeper understanding of the mechanisms regulating the SASP and components necessary for robust anti-tumor immune surveillance in different cancer and therapy contexts are key to harnessing senescence for tumor control. Here we draw a roadmap to modulate TIS and its immune-stimulating features for cancer immunotherapy.
Collapse
Affiliation(s)
- Loretah Chibaya
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jarin Snyder
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Marcus Ruscetti
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA; Immunology and Microbiology Program, University of Massachusetts Chan Medical School, Worcester, MA, USA; Cancer Center, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
17
|
Banerjee M, Yaddanapudi K, States JC. Zinc supplementation prevents mitotic accumulation in human keratinocyte cell lines upon environmentally relevant arsenic exposure. Toxicol Appl Pharmacol 2022; 454:116255. [PMID: 36162444 PMCID: PMC9683715 DOI: 10.1016/j.taap.2022.116255] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/09/2022] [Accepted: 09/20/2022] [Indexed: 12/01/2022]
Abstract
Disrupted cell cycle progression underlies the molecular pathogenesis of multiple diseases. Chronic exposure to inorganic arsenic (iAs) is a global health issue leading to multi-organ cancerous and non-cancerous diseases. Exposure to supratherapeutic concentrations of iAs causes cellular accumulation in G2 or M phase of the cell cycle in multiple cell lines by inducing cyclin B1 expression. It is not clear if iAs exposure at doses corresponding to serum levels of chronically exposed populations (∼100 nM) has any effect on cell cycle distribution. In the present study we investigated if environmentally relevant iAs exposure induced cell cycle disruption and mechanisms thereof employing two human keratinocyte cell lines (HaCaT and Ker-CT), flow cytometry, immunoblots and quantitative real-time PCR (qRT-PCR). iAs exposure (100 nM; 24 h) led to mitotic accumulation of cells in both cell lines, along with the stabilization of ANAPC11 ubiquitination targets cyclin B1 and securin, without affecting their steady state mRNA levels. This result suggested that induction of cyclin B1 and securin is modulated at the level of protein degradation. Moreover, zinc supplementation successfully prevented iAs-induced mitotic accumulation and stabilization of cyclin B1 and securin without affecting their mRNA levels. Together, these data suggest that environmentally relevant iAs exposure leads to mitotic accumulation possibly by displacing zinc from the RING finger subunit of anaphase promoting complex/cyclosome (ANAPC11), the cell cycle regulating E3 ubiquitin ligase. This early cell cycle disruptive effect of environmentally relevant iAs concentration could underpin the molecular pathogenesis of multiple diseases associated with chronic iAs exposure.
Collapse
Affiliation(s)
- Mayukh Banerjee
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA.
| | - Kavitha Yaddanapudi
- Immuno-Oncology Group, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA; Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA; Department of Microbiology/Immunology, University of Louisville, Louisville, KY, USA
| | - J Christopher States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| |
Collapse
|
18
|
Magnetomechanical Stress-Induced Colon Cancer Cell Growth Inhibition. JOURNAL OF NANOTHERANOSTICS 2022. [DOI: 10.3390/jnt3030010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The application of magnetomechanical stress in cells using internalized magnetic nanoparticles (MNPs) actuated by low-frequency magnetic fields has been attracting considerable interest in the field of cancer research. Recent developments prove that magnetomechanical stress can inhibit cancer cells’ growth. However, the MNPs’ type and the magnetic field’s characteristics are crucial parameters. Their variability allows multiple combinations, which induce specific biological effects. We previously reported the antiproliferative effects induced in HT29 colon cancer cells by static-magnetic-field (200 mT)-actuated spherical MNPs (100 nm). Herein, we show that similar growth inhibitory effects are induced in other colon cancer cell lines. The effect of magnetomechanical stress was also examined in the growth rate of tumor spheroids. Moreover, we examined the biological mechanisms involved in the observed cell growth inhibition. Under the experimental conditions employed, no cell death was detected by PI (propidium iodide) staining analysis. Flow cytometry and Western blotting revealed that G2/M cell cycle arrest might mediate the antiproliferative effects. Furthermore, MNPs were found to locate in the lysosomes, and a decreased number of lysosomes was detected in cells that had undergone magnetomechanical stress, implying that the mechanical activation of the internalized MNPs could induce lysosome membrane disruption. Of note, the lysosomal acidic conditions were proven to affect the MNPs’ magnetic properties, evidenced by vibrating sample magnetometry (VSM) analysis. Further research on the combination of the described magnetomechanical stress with lysosome-targeting chemotherapeutic drugs could lay the groundwork for the development of novel anticancer combination treatment schemes.
Collapse
|
19
|
Vaali-Mohammed MA, Abdulla MH, Matou-Nasri S, Eldehna WM, Meeramaideen M, Elkaeed EB, El-Watidy M, Alhassan NS, Alkhaya K, Al Obeed O. The Anticancer Effects of the Pro-Apoptotic Benzofuran-Isatin Conjugate (5a) Are Associated With p53 Upregulation and Enhancement of Conventional Chemotherapeutic Drug Efficiency in Colorectal Cancer Cell Lines. Front Pharmacol 2022; 13:923398. [PMID: 36046830 PMCID: PMC9421242 DOI: 10.3389/fphar.2022.923398] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/07/2022] [Indexed: 11/30/2022] Open
Abstract
The present study aimed to investigate in-depth a cytotoxic novel benzofuran-isatin conjugate (5a, 3-methyl-N'-(2-oxoindolin-3-ylidene)benzofuran-2-carbohydrazide) with promising potential anticancer activities in colorectal adenocarcinoma HT29 and metastatic colorectal cancer (CRC) SW620 cell lines. Thus, the primary cell events involved in tumorigenicity, tumor development, metastasis, and chemotherapy response were explored. Both CRC cell lines were exposed to different concentrations of Compound 5a and then subjected to real-time cell viability, migration, and invasion assays, colony formation and cytotoxicity assays, and flow cytometry for cell cycle analysis and apoptosis determination. Western blot and RT-qPCR were performed to assess the protein and transcript expression levels of epithelial-mesenchymal transition (EMT), cell cycle, and apoptosis markers. We showed that the Compound 5a treatment exhibited anticancer effects through inhibition of HT29 and SW620 cell viability, migration, and invasion, in a dose-dependent manner, which were associated with the upregulation of the tumor suppressor p53. Compound 5a also inhibited the colony formation ability of HT29 and SW620 cells and reversed EMT markers E-cadherin and N-cadherin expression. CRC cell exposure to Compound 5a resulted in a cell cycle arrest at the G1/G0 phase in HT29 cells and at the G2/M phase in SW620 cells, along with the downregulation of cyclin A1 expression, described to be involved in the S phase entry. Furthermore, Compound 5a-induced apoptosis was associated with the downregulation of the anti-apoptotic Bcl-xl marker, upregulation of pro-apoptotic Bax and cytochrome c markers, and increased mitochondrial outer membrane permeability, suggesting the involvement of mitochondria-dependent apoptosis pathway. In addition, the combination studies of Compound 5a with the main conventional chemotherapeutic drugs 5-fluorouracil, irinotecan, and oxaliplatin showed a more potent cytotoxic effect in both CRC cells than a single treatment. In conclusion, our findings described the interesting in vitro anticancer properties of Compound 5a, shown to have possible antitumor, antimetastatic, and pro-apoptotic activities, with the enhancement of the cytotoxic efficiency of conventional chemotherapeutic drugs. In vivo studies are requested to confirm the promising anticancer potential of Compound 5a for CRC therapy.
Collapse
Affiliation(s)
- Mansoor-Ali Vaali-Mohammed
- Colorectal Research Chair, Department of Surgery, King Saud University College of Medicine, Riyadh, Saudi Arabia
- Department of Zoology, Jamal Mohamed College (Autonomous), Affiliated to Bharathidasan University, Tiruchirappalli, India
| | - Maha-Hamadien Abdulla
- Colorectal Research Chair, Department of Surgery, King Saud University College of Medicine, Riyadh, Saudi Arabia
- *Correspondence: Maha-Hamadien Abdulla,
| | - Sabine Matou-Nasri
- King Abdullah International Medical Research Center, Cell and Gene Therapy Group, Medical Genomics Research Department, Ministry of National Guard Health Affairs, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Wagdy M. Eldehna
- School of Biotechnology, Badr University in Cairo, Badr, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - M. Meeramaideen
- Department of Zoology, Jamal Mohamed College (Autonomous), Affiliated to Bharathidasan University, Tiruchirappalli, India
| | - Eslam B. Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
| | - Mohammed El-Watidy
- College of Medicine Research Center (CMRC), King Saud University College of Medicine, Riyadh, Saudi Arabia
| | - Noura S. Alhassan
- Colorectal Research Chair, Department of Surgery, King Saud University College of Medicine, Riyadh, Saudi Arabia
| | - Khayal Alkhaya
- Colorectal Research Chair, Department of Surgery, King Saud University College of Medicine, Riyadh, Saudi Arabia
| | - Omar Al Obeed
- Colorectal Research Chair, Department of Surgery, King Saud University College of Medicine, Riyadh, Saudi Arabia
| |
Collapse
|
20
|
Ren Y, Gao F, Li B, Yuan A, Zheng L, Zhang Y. A precise efficacy determination strategy of traditional Chinese herbs based on Q-markers: Anticancer efficacy of Astragali radix as a case. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 102:154155. [PMID: 35580440 DOI: 10.1016/j.phymed.2022.154155] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND As a "multi-components and multi-efficacy" complex system, traditional Chinese herbs are universally distributed and applied in treating clinical diseases. However, the efficacy deviation and ambiguous clinical location are affected by different effects and content of components caused by uncertain factors in the production process. It further restricts resource allocation and clinical medication and hinders modernization and globalization. In this study, a precise efficacy determination strategy was innovatively proposed, aiming to quantitatively predict the efficacy of herbs and obtain precise medicinal materials. Quality-markers (Q-markers) characterizing the efficacy are conducive to achieving precise efficacy determination. PURPOSE With the anticancer efficacy of Astragali radix (AR) as a case, the present study was designed to establish a methodology for precise efficacy determination based on Q-markers characterizing specific efficacy. METHODS Guided by the basic principles of Q-markers, the potential Q-markers characterizing the anticancer efficacy of AR were screened through molecular simulation and network pharmacology. The activity of Q-markers was evaluated on MDA-MB-231 cells, and the content of Q-markers was determined by HPLC. A quantitative efficacy prediction model of the relationship between the influencing factors and anticancer efficacy was further constructed through the effect-constituents index (ECI) and machine learning and verified by biotechnology, which can be directly applied to predict the efficacy in numerous samples. RESULTS Astragaloside I, astragaloside II, and astragaloside III inhibited the proliferation of MDA-MB-231 cells and were successfully quantified in AR samples, reflecting the effectiveness and measurability of Q-markers. Gradient Boost Regression showed the best performance in the quantitative efficacy prediction model with EVtest= 0.815, R2test= 0.802. The results of precise efficacy determination indicated that 1-2-3 (Wuzhai, Shanxi, two years, C segment) sample performed best in 54 batches of AR samples with biased anticancer efficacy. Furthermore, AR samples with higher ECI had higher anticancer efficacy and vice versa. CONCLUSION The precise efficacy determination strategy established in the present study is reliable and proved in the AR case, which is expected to support resource allocation optimization, efficacy stability improvement, and precise clinical medication achievement.
Collapse
Affiliation(s)
- Yue Ren
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Fengfeng Gao
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Beiyan Li
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Anlei Yuan
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Lulu Zheng
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yanling Zhang
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
21
|
Mitochondrial micropeptide STMP1 promotes G1/S transition by enhancing mitochondrial complex IV activity. Mol Ther 2022; 30:2844-2855. [PMID: 35450818 PMCID: PMC9372290 DOI: 10.1016/j.ymthe.2022.04.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 02/18/2022] [Accepted: 04/18/2022] [Indexed: 12/24/2022] Open
Abstract
The roles of micropeptides in cell cycle regulation and cancer development remain largely unknown. Here we found that a micropeptide STMP1 (small transmembrane protein 1) was up-regulated in multiple malignancies including hepatocellular carcinoma (HCC), and its high level was associated with short recurrence-free survival of HCC patients. Gain- and loss-of-function analyses revealed that STMP1 accelerated cell proliferation and clonogenicity in vitro and tumor growth in vivo, and silencing STMP1 blocked G1/S transition. Mechanistically, STMP1 promoted the mRNA and protein levels of CCNE2, CDK2 and E2F1. STMP1 was localized in the inner membrane of mitochondria and interacted with mitochondrial complex IV and then enhanced its activity. Moreover, treatment with the mitochondrial complex IV inhibitor tetrathiomolybdate dramatically abrogated the promoting effect of STMP1 on cell proliferation and the expression of cyclin E2, CDK2 and E2F1. These results suggest that STMP1 may promote G1/S transition and cell proliferation by enhancing mitochondrial complex IV activity, which highlight STMP1 as a new regulator of the cell cycle and a potential target for anti-cancer therapy.
Collapse
|
22
|
Mazloum-Ravasan S, Mohammadi M, Hiagh EM, Ebrahimi A, Hong JH, Hamishehkar H, Kim KH. Nano-liposomal zein hydrolysate for improved apoptotic activity and therapeutic index in lung cancer treatment. Drug Deliv 2022; 29:1049-1059. [PMID: 35363101 PMCID: PMC8979517 DOI: 10.1080/10717544.2022.2057618] [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] [Indexed: 11/30/2022] Open
Abstract
Lung cancer is one of the most common cancers in the world with a high mortality rate. Zein is a protein compound whose protein isolate is not useful and whose protein hydrolysis produces biological activity. By encapsulating this bioactive compound inside the nanoparticles (NPs), it causes itself to reach the tumor site and destroy it rapidly. In this study, the effects of zein hydrolysate (ZH) and nano-liposomal ZH (N-ZH) were investigated on the human A549 cell line. Western blotting and cell cycle analyses showed that ZH and N-ZH caused cytotoxicity. They induced apoptosis via cell cycle arrest at the G0 phase, as well as significant increases in pro-apoptotic genes, such as Bax, caspase-3, -8, -9, and p53, accompanied with significant decreases in the anti-apoptotic marker Bcl-2. Based on the results, the cytotoxic and anticancer effects of N-ZH were higher than those of free ZH. In conclusion, liposomes improved the performance of ZH and dramatically reduced the IC50 value of ZH. These findings provided the experimental evidence that N-ZH with favorable anticancer activity can be used as a therapeutic agent and strategy for lung cancer treatment in future clinical trials.
Collapse
Affiliation(s)
| | - Maryam Mohammadi
- Department of Food Science and Engineering, Faculty of Agriculture, University of Kurdistan, Sanandaj, Iran.,Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elaheh Madadi Hiagh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.,Pediatrics III, University Hospital Essen, Essen, Germany
| | - Alireza Ebrahimi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Joo-Hyun Hong
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
23
|
Hu Y, Burkard ME. Classes of therapeutics to amplify the immune response. Breast Cancer Res Treat 2022; 191:277-289. [PMID: 34787761 PMCID: PMC11646685 DOI: 10.1007/s10549-021-06369-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/18/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Conventional chemotherapies are a mainstay for metastatic breast cancers, though durable response is rare. Immunotherapies promise long-term responses thorough immune activation but have been underwhelming in breast cancer relative to other cancer types. Here, we review the mechanisms of existing strategies including chemotherapies and how they may cause breast cancers to become immunogenic to identify potential biomarkers for combinations of conventional and immunotherapies. CONCLUSION Mechanistic considerations should inform biomarker development and patient selection for therapeutic combinations of drugs to combine with immune-checkpoint inhibitors.
Collapse
Affiliation(s)
- Yang Hu
- Department of Medicine, Hematology/Oncology and Palliative Care, University of Wisconsin-Madison, Madison, WI, 53705, USA
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, 53705, USA
- UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Medical Scientist Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Mark E Burkard
- Department of Medicine, Hematology/Oncology and Palliative Care, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Wisconsin Institutes for Medical Research, School of Medicine and Public Health, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
24
|
Abd El-wahab HA, Mansour HS, Ali AM, El-Awady R, Aboul-Fadl T. New Cell Cycle Checkpoint Pathways Regulators with 2-Oxo-indoline Scaffold as Potential Anticancer Agents: Design, Synthesis, Biological Activities and In Silico Studies. Bioorg Chem 2022; 120:105622. [DOI: 10.1016/j.bioorg.2022.105622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 12/18/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022]
|
25
|
Xiao Y, Dong J. The Hippo Signaling Pathway in Cancer: A Cell Cycle Perspective. Cancers (Basel) 2021; 13:cancers13246214. [PMID: 34944834 PMCID: PMC8699626 DOI: 10.3390/cancers13246214] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 01/25/2023] Open
Abstract
Simple Summary Cancer is increasingly viewed as a cell cycle disease in that the dysregulation of the cell cycle machinery is a common feature in cancer. The Hippo signaling pathway consists of a core kinase cascade as well as extended regulators, which together control organ size and tissue homeostasis. The aberrant expression of cell cycle regulators and/or Hippo pathway components contributes to cancer development, and for this reason, we specifically focus on delineating the roles of the Hippo pathway in the cell cycle. Improving our understanding of the Hippo pathway from a cell cycle perspective could be used as a powerful weapon in the cancer battlefield. Abstract Cell cycle progression is an elaborate process that requires stringent control for normal cellular function. Defects in cell cycle control, however, contribute to genomic instability and have become a characteristic phenomenon in cancers. Over the years, advancement in the understanding of disrupted cell cycle regulation in tumors has led to the development of powerful anti-cancer drugs. Therefore, an in-depth exploration of cell cycle dysregulation in cancers could provide therapeutic avenues for cancer treatment. The Hippo pathway is an evolutionarily conserved regulator network that controls organ size, and its dysregulation is implicated in various types of cancers. Although the role of the Hippo pathway in oncogenesis has been widely investigated, its role in cell cycle regulation has not been comprehensively scrutinized. Here, we specifically focus on delineating the involvement of the Hippo pathway in cell cycle regulation. To that end, we first compare the structural as well as functional conservation of the core Hippo pathway in yeasts, flies, and mammals. Then, we detail the multi-faceted aspects in which the core components of the mammalian Hippo pathway and their regulators affect the cell cycle, particularly with regard to the regulation of E2F activity, the G1 tetraploidy checkpoint, DNA synthesis, DNA damage checkpoint, centrosome dynamics, and mitosis. Finally, we briefly discuss how a collective understanding of cell cycle regulation and the Hippo pathway could be weaponized in combating cancer.
Collapse
Affiliation(s)
| | - Jixin Dong
- Correspondence: ; Tel.: +402-559-5596; Fax: +402-559-4651
| |
Collapse
|
26
|
da Costa Rosa M, Yamashita AS, Riggins GJ. Evaluation of a DNA demethylating agent in combination with all-trans retinoic acid for IDH1-mutant gliomas. Neuro Oncol 2021; 24:711-723. [PMID: 34850159 DOI: 10.1093/neuonc/noab263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Isocitrate Dehydrogenase 1/2 (IDH1/2) mutations are diagnostic for Astrocytoma or Oligodendroglioma, IDH-mutant. In these IDH-mutant gliomas, retinoic acid-related gene expression is commonly silenced by DNA hypermethylation. DNA demethylating agents can epigenetically reprogram IDH-mutant cells and reduce proliferation, likely by re-expression of silenced tumor suppressor pathways. We hypothesized that DNA demethylation might restore the retinoic acid pathway and slow tumor growth. This was the rationale for a preclinical evaluation combining the DNA demethylating agent, 5-Azacytidine (5-Aza), and retinoic acid pathway activation with all-trans retinoic acid (atRA) in IDH-mutant glioma. METHODS In this study, we evaluated the effect of 5-Aza and atRA combination on cell proliferation, apoptosis and gene expression in human glioma cells. In addition, the efficacy of combination was tested in patient-derived xenograft (PDX) bearing the IDH1R132H mutation, utilizing subcutaneous and orthotopic models. RESULTS 5-Aza reduced the DNA methylation profile and increased the gene expression of retinoic acid-related genes. Combination of 5-Aza and atRA reduced cell growth, increased differentiation marker expression, and apoptosis in IDH1R132H glioma cells. Mechanistically, 5-Aza sensitized IDHIR132H glioma cells to atRA via upregulation of the retinoic acid pathway. Importantly, the drug combination reduced significantly the growth rate of subcutaneous tumors, but in an orthotopic mouse model the combination did not improve survival and 5-Aza alone provided the best survival benefit. CONCLUSION Use of DNA demethylating agent in combination with retinoids shows promise, but further optimization and preclinical studies are required for treatment of intracranial IDH-mutant gliomas.
Collapse
Affiliation(s)
- Marina da Costa Rosa
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alex Shimura Yamashita
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gregory J Riggins
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
27
|
Yang Z, Yang M, Chow HM, Tsang SY, Lee MM, Chan MK. Cytosolic delivery of CDK4/6 inhibitor p16 protein using engineered protein crystals for cancer therapy. Acta Biomater 2021; 135:582-592. [PMID: 34496285 DOI: 10.1016/j.actbio.2021.08.054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/16/2021] [Accepted: 08/31/2021] [Indexed: 12/13/2022]
Abstract
The tumor suppressor p16 protein is an endogenous CDK4/6 inhibitor. Inactivation of its encoding gene is found in nearly half of human cancers. Restoration of p16 function via adenovirus-based gene delivery has been shown to be effective in suppressing aberrant cell growth in many types of cancer, however, the potential risk of insertional mutagenesis in genomic DNA remains a major concern. Thus, there has been great interest in developing efficient strategies to directly deliver proteins into cells as an alternative that can avoid such safety concerns while achieving a comparable therapeutic effect. Nevertheless, intracellular delivery of protein therapeutics remains a challenge. Our group has recently developed a protein delivery platform based on an engineered Pos3Aa protein that forms sub-micrometer-sized crystals in Bacillus thuringiensis cells. In this report, we describe the further development of this platform (Pos3AaTM) via rationally designed site-directed mutagenesis, and its resultant potency for the delivery of cargo proteins into cells. Pos3AaTM-based fusion protein crystals are shown to exhibit improved release of their cargo proteins as demonstrated using a model mCherry protein. Importantly, this Pos3AaTM platform is able to mediate the efficient intracellular delivery of p16 protein with significant endosomal escape, resulting in p16-mediated inhibition of CDK4/6 kinase activity and Rb phosphorylation, and as a consequence, significant cell cycle arrest and cell growth inhibition. These results validate the ability of these improved Pos3AaTM crystals to mediate enhanced cytosolic protein delivery and highlight the potential of using protein therapeutics as selective CDK4/6 inhibitors for cancer therapy. STATEMENT OF SIGNIFICANCE: Cytosolic delivery of bioactive therapeutic proteins capable of eliciting therapeutic benefit remains a significant challenge. We have previously developed a protein delivery platform based on engineered Pos3Aa protein crystals with excellent cell-permeability and endosomal escape properties. In this report, we describe the rational design of an improved Pos3Aa triple mutant (Pos3AaTM) with enhanced cargo release. We demonstrate that Pos3AaTM-mCherry-p16 fusion crystals can efficiently deliver p16 protein, a CDK4/6 inhibitor frequently inactivated in human cancers, into p16-deficient UM-SCC-22A cells, where it promotes significant G1 cell cycle arrest and cell growth inhibition. These results highlight the ability of the Pos3AaTM platform to promote potent cytosolic delivery of protein therapeutics, and the efficacy of p16 protein delivery as an effective strategy for treating cancer.
Collapse
Affiliation(s)
- Zaofeng Yang
- School of Life Sciences and Center of Novel Biomaterials, The Chinese University of Hong Kong, China Hong Kong Special Administrative Region
| | - Meigui Yang
- School of Life Sciences and Center of Novel Biomaterials, The Chinese University of Hong Kong, China Hong Kong Special Administrative Region
| | - Hei-Man Chow
- School of Life Sciences and Center of Novel Biomaterials, The Chinese University of Hong Kong, China Hong Kong Special Administrative Region
| | - Suk Ying Tsang
- School of Life Sciences and Center of Novel Biomaterials, The Chinese University of Hong Kong, China Hong Kong Special Administrative Region; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, China Hong Kong Special Administrative Region
| | - Marianne M Lee
- School of Life Sciences and Center of Novel Biomaterials, The Chinese University of Hong Kong, China Hong Kong Special Administrative Region.
| | - Michael K Chan
- School of Life Sciences and Center of Novel Biomaterials, The Chinese University of Hong Kong, China Hong Kong Special Administrative Region.
| |
Collapse
|
28
|
A machine learning approach for single cell interphase cell cycle staging. Sci Rep 2021; 11:19278. [PMID: 34588507 PMCID: PMC8481278 DOI: 10.1038/s41598-021-98489-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 09/09/2021] [Indexed: 11/30/2022] Open
Abstract
The cell nucleus is a tightly regulated organelle and its architectural structure is dynamically orchestrated to maintain normal cell function. Indeed, fluctuations in nuclear size and shape are known to occur during the cell cycle and alterations in nuclear morphology are also hallmarks of many diseases including cancer. Regrettably, automated reliable tools for cell cycle staging at single cell level using in situ images are still limited. It is therefore urgent to establish accurate strategies combining bioimaging with high-content image analysis for a bona fide classification. In this study we developed a supervised machine learning method for interphase cell cycle staging of individual adherent cells using in situ fluorescence images of nuclei stained with DAPI. A Support Vector Machine (SVM) classifier operated over normalized nuclear features using more than 3500 DAPI stained nuclei. Molecular ground truth labels were obtained by automatic image processing using fluorescent ubiquitination-based cell cycle indicator (Fucci) technology. An average F1-Score of 87.7% was achieved with this framework. Furthermore, the method was validated on distinct cell types reaching recall values higher than 89%. Our method is a robust approach to identify cells in G1 or S/G2 at the individual level, with implications in research and clinical applications.
Collapse
|
29
|
Mumblat H, Martinez-Conde A, Braten O, Munster M, Dor-On E, Schneiderman RS, Porat Y, Voloshin T, Davidi S, Blatt R, Shteingauz A, Tempel-Brami C, Zeevi E, Lajterer C, Shmueli Y, Danilov S, Haber A, Giladi M, Weinberg U, Kinzel A, Palti Y. Tumor Treating Fields (TTFields) downregulate the Fanconi Anemia-BRCA pathway and increase the efficacy of chemotherapy in malignant pleural mesothelioma preclinical models. Lung Cancer 2021; 160:99-110. [PMID: 34482104 DOI: 10.1016/j.lungcan.2021.08.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/19/2021] [Accepted: 08/21/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVES Tumor Treating Fields (TTFields) are low intensity, intermediate frequency, alternating electric fields with antimitotic effects on cancerous cells. TTFields concomitant with pemetrexed and a platinum agent are approved in the US and EU as first line therapy for unresectable, locally advanced or metastatic malignant pleural mesothelioma (MPM). The goal of the current study was to characterize the mechanism of action of TTFields in MPM cell lines and animal models. METHODS Human MPM cell lines MSTO-211H and NCI-H2052 were treated with TTFields to determine the frequency that elicits maximal cytotoxicity. The effect of TTFields on DNA damage and repair, and the cytotoxic effect of TTFields in combination with cisplatin and/or pemetrexed were examined. Efficacy of TTFields concomitant with cisplatin and pemetrexed was evaluated in orthotopic IL-45 and subcutaneous RN5 murine models. RESULTS TTFields at a frequency of 150 kHz demonstrated the highest cytotoxicity to MPM cells. Application of 150 kHz TTFields resulted in increased formation of DNA double strand breaks, elevated expression of DNA damage induced cell cycle arrest proteins, and reduced expression of Fanconi Anemia (FA)-BRCA DNA repair pathway proteins. Co-treatment of TTFields with cisplatin or pemetrexed significantly increased treatment efficacy versus each modality alone, with additivity and synergy exhibited by the TTFields-pemetrexed and TTFields-cisplatin combinations, respectively. In animal models, tumor volume was significantly lower for the TTFields-cisplatin-pemetrexed combination compared to control, accompanied by increased DNA damage within the tumor. CONCLUSION This research demonstrated that the efficacy of TTFields for the treatment of MPM is associated with reduced expression of FA-BRCA pathway proteins and increased DNA damage. This mechanism of action is consistent with the observed synergism for TTFields-cisplatin vs additivity for TTFields-pemetrexed, as cisplatin-induced DNA damage is repaired via the FA-BRCA pathway.
Collapse
|
30
|
Simic M, Petkovic M, Jovanovic P, Jovanovic M, Tasic G, Besu I, Zizak Z, Aleksic I, Nikodinovic-Runic J, Savic V. Fragment-type 4-azolylcoumarin derivatives with anticancer properties. Arch Pharm (Weinheim) 2021; 354:e2100238. [PMID: 34374111 DOI: 10.1002/ardp.202100238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/23/2022]
Abstract
Several coumarin derivatives with a directly attached azole substituent at C-4 were synthesized and biologically studied for their anticancer properties. The cell lines used for this investigation (HeLa, K-562, MDA-MB-53, and MCF-7) demonstrated different sensitivities. The best response in the MTT (3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide) assay was shown by K-562 cells, with compounds displaying activity (3c, IC50 3.06 μM; 4a, IC50 5.24 μM; 4c, IC50 4.7 μM) similar to that of cisplatin (IC50 ~6 μM), which was used as the standard. The studied azole-substituted coumarins demonstrated weaker activity toward other cell lines, except for compound 4c, which was equally potent in the case of MCF-7 cells. Additional biological evaluations supported interference with the cell cycle as a potential mechanism of action and confirmed the absence of toxicity in zebrafish embryos. On the basis of these initial results, 4-azole coumarins should be explored further. Although their activity would need additional optimization, the fact that these compounds are fragment-like structures with MW <300 and clog P <3 offers enough flexibility to fine-tune their drug-like properties.
Collapse
Affiliation(s)
- Milena Simic
- Department of Organic Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Milos Petkovic
- Department of Organic Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Predrag Jovanovic
- Department of Organic Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Milos Jovanovic
- Department of Organic Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Gordana Tasic
- Department of Organic Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Irina Besu
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Zeljko Zizak
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Ivana Aleksic
- Laboratory for Eco-Biotechnology and Drug Development, Institute for Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Jasmina Nikodinovic-Runic
- Laboratory for Eco-Biotechnology and Drug Development, Institute for Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Vladimir Savic
- Department of Organic Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
31
|
Peng Y, Tao H, Gao Y, Yang Y, Chen Z. Review and Prospect of Tissue-agnostic Targeted Strategies in Anticancer Therapies. Curr Top Med Chem 2021; 21:404-425. [PMID: 32543358 DOI: 10.2174/1568026620666200616143247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/04/2020] [Accepted: 05/08/2020] [Indexed: 11/22/2022]
Abstract
Due to the increasing prevalence of cancer year by year, and the complexity and refractory nature of the disease itself, it is required to constantly innovate the development of new cancer treatment schemes. At the same time, the understanding of cancers has deepened, from the use of chemotherapy regimens with high toxicity and side effects, to the popularity of targeted drugs with specific targets, to precise treatments based on tumor characteristics rather than traditional anatomical location classification. In precision medicine, in the view of the specific cancer diseases and their biological characteristics, there is a great potential to develop tissue-agnostic targeted therapy with broad-spectrum anticancer significance. The present review has discussed tissue-agnostic targeted therapy based on the biological and genetic characteristics of cancers, expounded its theoretical basis and strategies for drug development. In addition, the feasible drug targets, FDA-approved drugs, as well as drug candidates in clinical trials have also been summarized. In conclusion, the "tissue-agnostic targeted therapy" is a breakthrough in anticancer therapies.
Collapse
Affiliation(s)
- Yu Peng
- Jiangsu Provincial Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Hongxun Tao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Yuanqing Gao
- Jiangsu Provincial Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yuanyuan Yang
- Xi'an Institute for Food and Drug Control, Xi'an Shaanxi 710054, China
| | - Zhiyong Chen
- Shaanxi Academy of Traditional Chinese Medicine, Xi'an Shaanxi 710003, China
| |
Collapse
|
32
|
Cohen P, Cross D, Jänne PA. Kinase drug discovery 20 years after imatinib: progress and future directions. Nat Rev Drug Discov 2021; 20:551-569. [PMID: 34002056 PMCID: PMC8127496 DOI: 10.1038/s41573-021-00195-4] [Citation(s) in RCA: 540] [Impact Index Per Article: 180.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2021] [Indexed: 02/04/2023]
Abstract
Protein kinases regulate nearly all aspects of cell life, and alterations in their expression, or mutations in their genes, cause cancer and other diseases. Here, we review the remarkable progress made over the past 20 years in improving the potency and specificity of small-molecule inhibitors of protein and lipid kinases, resulting in the approval of more than 70 new drugs since imatinib was approved in 2001. These compounds have had a significant impact on the way in which we now treat cancers and non-cancerous conditions. We discuss how the challenge of drug resistance to kinase inhibitors is being met and the future of kinase drug discovery.
Collapse
Affiliation(s)
- Philip Cohen
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK.
| | | | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana Farber Cancer Institute, Harvard University, Boston, MA, USA.
| |
Collapse
|
33
|
Vitamin E TPGS 1000 Induces Apoptosis in the K562 Cell Line: Implications for Chronic Myeloid Leukemia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5580288. [PMID: 34211630 PMCID: PMC8211508 DOI: 10.1155/2021/5580288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/14/2021] [Accepted: 05/20/2021] [Indexed: 11/17/2022]
Abstract
Chronic myeloid leukemia (CML) is a hematologic malignancy derived from the myeloid lineage molecularly characterized by t(9;22)(q34;q11) resulting in BCR-ABL1 gene fusion, which is known as Philadelphia (Ph) chromosome. Although tyrosine kinase inhibitors (TKIs) have restored and maintained the quality of life of patients with CML, an important minority of patients become resistant to first-and-second-generation TKIs and require an alternative treatment. The K562 cell (Ph+, p53-/-) line was treated with Vit E TPGS 1000 (20-80 μM) only or with other products of interest (e.g., antioxidant N-acetylcysteine (NAC), specific JNK and caspase-3 inhibitor SP600125, and NSCSI, respectively) for 24 h at 37°C. Cells were analyzed by fluorescence microscopy (FM), flow cytometry (FC), and Western blotting (WB) techniques. We show that TPGS induces apoptosis in K562 cells through H2O2 signaling mechanism comprising the activation of a minimal molecular cascade: the kinase JNK>the transcription factor c-JUN>the activation of BCL-only BH3 proapoptotic protein PUMA>loss of mitochondrial membrane potential (ΔΨ m)>activation of caspase-3>chromatin condensation>fragmentation of DNA. Additionally, TPGS oxidizes the stress sensor protein DJ-1-Cys106-SH into DJ-1-Cys106-SO3 and arrested the cell cycle in the S phase. Remarkably, NAC, SP600125, and NSCSI blocked TPGS-induced OS and apoptosis in K562. Since TPGS is safe in mice and humans, it is especially promising for preclinical and clinical CML leukemia research. Our findings support the view that oxidation therapy offers an important opportunity to eliminate CML.
Collapse
|
34
|
Biayna J, Garcia-Cao I, Álvarez MM, Salvadores M, Espinosa-Carrasco J, McCullough M, Supek F, Stracker TH. Loss of the abasic site sensor HMCES is synthetic lethal with the activity of the APOBEC3A cytosine deaminase in cancer cells. PLoS Biol 2021; 19:e3001176. [PMID: 33788831 PMCID: PMC8041192 DOI: 10.1371/journal.pbio.3001176] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 04/12/2021] [Accepted: 03/08/2021] [Indexed: 12/26/2022] Open
Abstract
Analysis of cancer mutagenic signatures provides information about the origin of mutations and can inform the use of clinical therapies, including immunotherapy. In particular, APOBEC3A (A3A) has emerged as a major driver of mutagenesis in cancer cells, and its expression results in DNA damage and susceptibility to treatment with inhibitors of the ATR and CHK1 checkpoint kinases. Here, we report the implementation of CRISPR/Cas-9 genetic screening to identify susceptibilities of multiple A3A-expressing lung adenocarcinoma (LUAD) cell lines. We identify HMCES, a protein recently linked to the protection of abasic sites, as a central protein for the tolerance of A3A expression. HMCES depletion results in synthetic lethality with A3A expression preferentially in a TP53-mutant background. Analysis of previous screening data reveals a strong association between A3A mutational signatures and sensitivity to HMCES loss and indicates that HMCES is specialized in protecting against a narrow spectrum of DNA damaging agents in addition to A3A. We experimentally show that both HMCES disruption and A3A expression increase susceptibility of cancer cells to ionizing radiation (IR), oxidative stress, and ATR inhibition, strategies that are often applied in tumor therapies. Overall, our results suggest that HMCES is an attractive target for selective treatment of A3A-expressing tumors.
Collapse
Affiliation(s)
- Josep Biayna
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Isabel Garcia-Cao
- Genomic Instability and Cancer, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Miguel M. Álvarez
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Marina Salvadores
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Jose Espinosa-Carrasco
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Marcel McCullough
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Fran Supek
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- * E-mail: (FS); (THS)
| | - Travis H. Stracker
- Genomic Instability and Cancer, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- National Cancer Institute, Center for Cancer Research, Radiation Oncology Branch, Bethesda, Maryland, United States of America
- * E-mail: (FS); (THS)
| |
Collapse
|
35
|
Mallela K, Kumar A. Role of TSC1 in physiology and diseases. Mol Cell Biochem 2021; 476:2269-2282. [PMID: 33575875 DOI: 10.1007/s11010-021-04088-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/27/2021] [Indexed: 12/15/2022]
Abstract
Since its initial discovery as the gene altered in Tuberous Sclerosis Complex (TSC), an autosomal dominant disorder, the interest in TSC1 (Tuberous Sclerosis Complex 1) has steadily risen. TSC1, an essential component of the pro-survival PI3K/AKT/MTOR signaling pathway, plays an important role in processes like development, cell growth and proliferation, survival, autophagy and cilia development by co-operating with a variety of regulatory molecules. Recent studies have emphasized the tumor suppressive role of TSC1 in several human cancers including liver, lung, bladder, breast, ovarian, and pancreatic cancers. TSC1 perceives inputs from various signaling pathways, including TNF-α/IKK-β, TGF-β-Smad2/3, AKT/Foxo/Bim, Wnt/β-catenin/Notch, and MTOR/Mdm2/p53 axis, thereby regulating cancer cell proliferation, metabolism, migration, invasion, and immune regulation. This review provides a first comprehensive evaluation of TSC1 and illuminates its diverse functions apart from its involvement in TSC genetic disorder. Further, we have summarized the physiological functions of TSC1 in various cellular events and conditions whose dysregulation may lead to several pathological manifestations including cancer.
Collapse
Affiliation(s)
- Karthik Mallela
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | - Arun Kumar
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
36
|
Yi J, Li M, Yang M, Cai S, Zhao T, Cao J, Cheng G. Characterisation and
in vitro
cytotoxicity of toxic and degradation compounds in bamboo shoots (
Dendrocalamus Sinicus
) during traditional fermentation. Int J Food Sci Technol 2021. [DOI: 10.1111/ijfs.14750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Junjie Yi
- Faculty of Agriculture and Food Kunming University of Science and Technology Kunming650500China
- Yunnan Institute of Food Safety Kunming University of Science and Technology Kunming650500China
| | - Meiqi Li
- Faculty of Agriculture and Food Kunming University of Science and Technology Kunming650500China
- Yunnan Institute of Food Safety Kunming University of Science and Technology Kunming650500China
| | - Meilian Yang
- Faculty of Agriculture and Food Kunming University of Science and Technology Kunming650500China
- Yunnan Institute of Food Safety Kunming University of Science and Technology Kunming650500China
| | - Shengbao Cai
- Faculty of Agriculture and Food Kunming University of Science and Technology Kunming650500China
- Yunnan Institute of Food Safety Kunming University of Science and Technology Kunming650500China
| | - Tianrui Zhao
- Faculty of Agriculture and Food Kunming University of Science and Technology Kunming650500China
- Yunnan Institute of Food Safety Kunming University of Science and Technology Kunming650500China
| | - Jianxin Cao
- Faculty of Agriculture and Food Kunming University of Science and Technology Kunming650500China
- Yunnan Institute of Food Safety Kunming University of Science and Technology Kunming650500China
| | - Guiguang Cheng
- Faculty of Agriculture and Food Kunming University of Science and Technology Kunming650500China
- Yunnan Institute of Food Safety Kunming University of Science and Technology Kunming650500China
| |
Collapse
|
37
|
Lonati L, Barbieri S, Guardamagna I, Ottolenghi A, Baiocco G. Radiation-induced cell cycle perturbations: a computational tool validated with flow-cytometry data. Sci Rep 2021; 11:925. [PMID: 33441727 PMCID: PMC7806866 DOI: 10.1038/s41598-020-79934-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022] Open
Abstract
Cell cycle progression can be studied with computational models that allow to describe and predict its perturbation by agents as ionizing radiation or drugs. Such models can then be integrated in tools for pre-clinical/clinical use, e.g. to optimize kinetically-based administration protocols of radiation therapy and chemotherapy. We present a deterministic compartmental model, specifically reproducing how cells that survive radiation exposure are distributed in the cell cycle as a function of dose and time after exposure. Model compartments represent the four cell-cycle phases, as a function of DNA content and time. A system of differential equations, whose parameters represent transition rates, division rate and DNA synthesis rate, describes the temporal evolution. Initial model inputs are data from unexposed cells in exponential growth. Perturbation is implemented as an alteration of model parameters that allows to best reproduce cell-cycle profiles post-irradiation. The model is validated with dedicated in vitro measurements on human lung fibroblasts (IMR90). Cells were irradiated with 2 and 5 Gy with a Varian 6 MV Clinac at IRCCS Maugeri. Flow cytometry analysis was performed at the RadBioPhys Laboratory (University of Pavia), obtaining cell percentages in each of the four phases in all studied conditions up to 72 h post-irradiation. Cells show early \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${\text{G}}_{2}$$\end{document}G2-phase block (increasing in duration as dose increases) and later \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${\text{G}}_{1}$$\end{document}G1-phase accumulation. For each condition, we identified the best sets of model parameters that lead to a good agreement between model and experimental data, varying transition rates from \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${\text{G}}_{1}$$\end{document}G1- to S- and from \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${\text{G}}_{2}$$\end{document}G2- to M-phase. This work offers a proof-of-concept validation of the new computational tool, opening to its future development and, in perspective, to its integration in a wider framework for clinical use.
Collapse
Affiliation(s)
- Leonardo Lonati
- Radiation Biophysics and Radiobiology Group, Physics Department, University of Pavia, 27100, Pavia, IT, Italy.
| | - Sofia Barbieri
- Radiation Biophysics and Radiobiology Group, Physics Department, University of Pavia, 27100, Pavia, IT, Italy.,Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211, Geneva, CH, Switzerland
| | - Isabella Guardamagna
- Radiation Biophysics and Radiobiology Group, Physics Department, University of Pavia, 27100, Pavia, IT, Italy
| | - Andrea Ottolenghi
- Radiation Biophysics and Radiobiology Group, Physics Department, University of Pavia, 27100, Pavia, IT, Italy
| | - Giorgio Baiocco
- Radiation Biophysics and Radiobiology Group, Physics Department, University of Pavia, 27100, Pavia, IT, Italy
| |
Collapse
|
38
|
Lin WS, Leland JV, Ho CT, Pan MH. Occurrence, Bioavailability, Anti-inflammatory, and Anticancer Effects of Pterostilbene. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:12788-12799. [PMID: 32064876 DOI: 10.1021/acs.jafc.9b07860] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Supplementation with natural compounds found in fruits and vegetables has long been associated with a reduced risk of several types of cancer. Pterostilbene is a natural stilbenoid and a dimethylated analogue of resveratrol which is found primarily in blueberries. Pterostilbene exhibits a range of pharmacological properties, particularly anti-inflammatory and anticancer effects. Due to two methoxy groups in its skeleton, pterostilbene is more lipophilic than resveratrol and thus possesses higher intestinal permeability and cellular uptake and enhanced stability. Moreover, pterostilbene exhibits less toxicity and fewer adverse effects, providing it with superior potential in cancer chemoprevention and chemotherapy applications. Numerous research studies have demonstrated that pterostilbene possesses detoxification activities, mediating the anti-inflammation response, regulating the cell cycle, augmenting apoptosis, enhancing autophagy, and inhibiting tumor angiogenesis, invasion, and metastasis by modulating signal transduction pathways which block multiple stages of carcinogenesis. In this review, we illustrate that pterostilbene is a natural compound having bioavailability. The extensive metabolism of pterostilbene will be discussed. We also summarize recent research on pterostilbene's anti-inflammatory and anticancer properties in the multistage carcinogenesis process and related molecular mechanism and conclude that it should contribute to improved cancer management.
Collapse
Affiliation(s)
- Wei-Sheng Lin
- Institute of Food Science and Technology, National Taiwan University, Taipei 10617, Taiwan
| | | | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, New Jersey 08901-8520, United States
| | - Min-Hsiung Pan
- Institute of Food Science and Technology, National Taiwan University, Taipei 10617, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
39
|
Fluorinated benzylidene indanone exhibits antiproliferative activity through modulation of microtubule dynamics and antiangiogenic activity. Eur J Pharm Sci 2020; 154:105513. [PMID: 32805425 DOI: 10.1016/j.ejps.2020.105513] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 08/11/2020] [Accepted: 08/11/2020] [Indexed: 02/08/2023]
Abstract
The application of fluorine in drug design has been understood significantly by the medicinal chemists in recent years. Modulation of tubulin-microtubule dynamics is one of the most effective targets for cancer chemotherapeutics. A logically designed and identified lead compound, fluorinated benzylidene indanone 1 has been extensively evaluated for cancer pharmacology. It occupied colchicine binding pocket acting as microtubule destabilizer and induced a G2/M phase arrest in MCF-7 cells. Compound 1 exerted an antiangiogenic effect in MCF-7 cells by down-regulating Vascular Endothelial Growth Factor (VEGF) and Hypoxia Inducible Factor-α (HIF-α). In in-vivo efficacy in C3H/Jax mice mammary carcinoma model, benzylidene indanone 1 reduced tumour volumes by 48.2%. Further in acute oral toxicity studies compound 1 was well tolerated and safe up to 1000 mg/kg dose in Swiss albino mice. The fluorinated benzylidene indanone 1, a new chemical entity (NCE) can further be optimized for better efficacy against breast adenocarcinoma.1.
Collapse
|
40
|
Yamashita AS, da Costa Rosa M, Stumpo V, Rais R, Slusher BS, Riggins GJ. The glutamine antagonist prodrug JHU-083 slows malignant glioma growth and disrupts mTOR signaling. Neurooncol Adv 2020; 3:vdaa149. [PMID: 33681764 PMCID: PMC7920530 DOI: 10.1093/noajnl/vdaa149] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Metabolic reprogramming is a common feature in cancer, and it is critical to facilitate cancer cell growth. Isocitrate Dehydrogenase 1/2 (IDH1 and IDH2) mutations (IDHmut) are the most common genetic alteration in glioma grade II and III and secondary glioblastoma and these mutations increase reliance on glutamine metabolism, suggesting a potential vulnerability. In this study, we tested the hypothesis that the brain penetrant glutamine antagonist prodrug JHU-083 reduces glioma cell growth. Material and Methods We performed cell growth, cell cycle, and protein expression in glutamine deprived or Glutaminase (GLS) gene silenced glioma cells. We tested the effect of JHU-083 on cell proliferation, metabolism, and mTOR signaling in cancer cell lines. An orthotopic IDH1R132H glioma model was used to test the efficacy of JHU-083 in vivo. Results Glutamine deprivation and GLS gene silencing reduced glioma cell proliferation in vitro in glioma cells. JHU-083 reduced glioma cell growth in vitro, modulated cell metabolism, and disrupted mTOR signaling and downregulated Cyclin D1 protein expression, through a mechanism independent of TSC2 modulation and glutaminolysis. IDH1R132H isogenic cells preferentially reduced cell growth and mTOR signaling downregulation. In addition, guanine supplementation partially rescued IDHmut glioma cell growth, mTOR signaling, and Cyclin D1 protein expression in vitro. Finally, JHU-083 extended survival in an intracranial IDH1 mut glioma model and reduced intracranial pS6 protein expression. Conclusion Targeting glutamine metabolism with JHU-083 showed efficacy in preclinical models of IDHmut glioma and measurably decreased mTOR signaling.
Collapse
Affiliation(s)
- Alex Shimura Yamashita
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marina da Costa Rosa
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vittorio Stumpo
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rana Rais
- Johns Hopkins Drug Discovery, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gregory J Riggins
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
41
|
Yousef MA, Ali AM, El-Sayed WM, Qayed WS, Farag HHA, Aboul-Fadl T. Design and synthesis of novel isatin-based derivatives targeting cell cycle checkpoint pathways as potential anticancer agents. Bioorg Chem 2020; 105:104366. [PMID: 33212312 DOI: 10.1016/j.bioorg.2020.104366] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/08/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022]
Abstract
In recent years, cell cycle and checkpoint pathways regulation are offering new therapeutic approaches against cancer. Isatin, is a well exploited scaffold in the anticancer domain. Accordingly, the current work describes the design and synthesis of two series of (Z)-3-substituted-2-(((E/Z)-5-substituted-2-oxo-1-substituted-indolin-3-ylidene)hydrazinylidene)-thiazolidin-4-ones, 4(a-s) and (E/Z)-1-substituted-3-(((Z)-3-substituted-4-methylthiazol-2(3H)-ylidene)hydrazineylidene)-5-substituted-indolin-2-ones, 5(a-s). The structures of the synthesized molecules were confirmed by spectral and elemental methods of analyses. Pure diastereomers were further identified with 1H-1H-NOESY and confirmed with X-ray crystallography. The target compounds were tested in vitro for their cytotoxicity against three human epithelial cell lines, liver (HepG2), breast (MCF-7), and colon (HT-29) in addition to the diploid human normal cells (WI-38) compared to doxorubicin as a reference drug. Variable cytotoxic effects (IC50 3.29-100 µmol) were reported on the three cancer cell lines with pronounced selectivity compared to the normal one WI-38. The potency of the most active compounds, 4o, 4s, 5e, 5f, 5l, 5m and 5o (IC50 3.29-9.92 µmol), in both series associated with the (Z) configurations of N = thiazolidin/ene or one, however, the configuration of the N = isatin moiety seemed to be of no importance to the activity. The tested compounds were grouped for their possible mechanism of action into 4 categories. Compound 4o with no apparent effect on all genes examined. Compounds 4s and 5o affected all genes investigated and seem to have multiple cellular targets; induced the expression of p53 and caspases, and downregulated that of CDK1. Compounds 5l and 5m directly elevated the expression of initiator and effector caspases without going through p53 pathway. Finally, compounds 5e and 5f elevated the expression of p53 and inhibited CDK1. Compounds 4s, 5e, 5f, 5l, 5m, and 5o caused a significant elevation in the activity of cleaved caspase 3 as well. Docking studies on CDK1 revealed that the active molecules bind to the tested enzyme by the same manner of the co-crystallized ligands and the isatin-thiazoldinone/ene scaffold is essential for binding of these molecules.
Collapse
Affiliation(s)
- Mohamed A Yousef
- Department of Medicinal Chemistry, Faculty of Pharmacy, Assuit University, Assuit 71526, Egypt
| | - Ahmed M Ali
- Department of Medicinal Chemistry, Faculty of Pharmacy, Assuit University, Assuit 71526, Egypt
| | - Wael M El-Sayed
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia 11566, Cairo, Egypt
| | - Wesam S Qayed
- Department of Medicinal Chemistry, Faculty of Pharmacy, Assuit University, Assuit 71526, Egypt.
| | - Hassan H A Farag
- Department of Medicinal Chemistry, Faculty of Pharmacy, Assuit University, Assuit 71526, Egypt
| | - Tarek Aboul-Fadl
- Department of Medicinal Chemistry, Faculty of Pharmacy, Assuit University, Assuit 71526, Egypt.
| |
Collapse
|
42
|
Nasr FA, Shahat AA, Alqahtani AS, Ahmed MZ, Qamar W, Al-Mishari AA, Almoqbil AN. Centaurea bruguierana inhibits cell proliferation, causes cell cycle arrest, and induces apoptosis in human MCF-7 breast carcinoma cells. Mol Biol Rep 2020; 47:6043-6051. [PMID: 32700127 DOI: 10.1007/s11033-020-05679-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 07/19/2020] [Indexed: 12/24/2022]
Abstract
Centaurea bruguierana, of the Asteraceae family, has a long history of use in traditional medicines for the treatment of various ailments. However, the anticancer activity and underlying mechanisms have not yet been assessed. The C. bruguierana was extracted with methanol and fractionated into four different fractions. Different cancer cells and one non-cancerous were used to examine the cytotoxic effects of these fractions using MTT assay. The most potent fraction, C. bruguierana ethyl acetate fraction (CB EtOAc), was explored for its effects on cell cycle progression and apoptosis induction by Hoechst staining and annexin V-PI double staining in MCF-7 cells. The expression of apoptosis-related genes was quantified by RT-PCR. Of all fractions, CB EtOAc was found to have the strongest antiproliferative activity (IC50 = 10 μg/mL) against MCF-7 cells. The antiproliferative activity of the CB EtOAc fraction against MCF-7 was correlated with arrested of cell cycle in the G1 phase, nuclear fragmentation, and the exposure of phosphatidylserine. The induction of apoptosis by CB EtOAc in MCF-7 cells was also associated with an increase in the Bax/Bcl-2 ratio and higher expression of caspases. Overall, our results demonstrated that CB EtOAc showed apoptosis-inducing effects, suggesting that C. bruguierana may be a promising source for a novel chemotherapeutic agents for the treatment of breast cancer.
Collapse
Affiliation(s)
- Fahd A Nasr
- Medicinal, Aromatic and Poisonous Plants Research Center, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia.
| | - Abdelaaty A Shahat
- Medicinal, Aromatic and Poisonous Plants Research Center, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia.,Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia.,Chemistry of Medicinal Plants Dept., National Research Centre, 33 El Bohouth St. (former El Tahrirst.), Dokki, P. O. 12622, Giza, Egypt
| | - Ali S Alqahtani
- Medicinal, Aromatic and Poisonous Plants Research Center, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia.,Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Mohammad Z Ahmed
- Medicinal, Aromatic and Poisonous Plants Research Center, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia.,Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Wajhul Qamar
- Central Laboratory, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Abdullah A Al-Mishari
- Medicinal, Aromatic and Poisonous Plants Research Center, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Abdulaziz N Almoqbil
- Medicinal, Aromatic and Poisonous Plants Research Center, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
43
|
Song M, Lan Y, Wu X, Han Y, Wang M, Zheng J, Li Z, Li F, Zhou J, Xiao J, Cao Y, Xiao H. The chemopreventive effect of 5-demethylnobiletin, a unique citrus flavonoid, on colitis-driven colorectal carcinogenesis in mice is associated with its colonic metabolites. Food Funct 2020; 11:4940-4952. [PMID: 32459257 PMCID: PMC10726105 DOI: 10.1039/d0fo00616e] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
5-Demethylnobiletin (5DN) is a unique flavonoid mainly found in citrus fruits. In this study, we determined the chemopreventive effects of 5DN and its major colonic metabolites on both a colitis-driven colon carcinogenesis mouse model and a human colon cancer cell model. In azoxymethane/dextran sulfate sodium-treated mice, dietary 5DN (0.05% w/w in the diet) significantly decreased the tumor incidence, multiplicity and burden, and showed potent anti-proliferative, proapoptotic, and anti-inflammatory activities in mouse colon tissue. Three major metabolites of 5DN, named 5,3'-didemethylnobiletin (M1), 5,4'-didemethylnobiletin (M2) and 5,3',4'-tridemethylnobiletin (M3), were found in the colonic mucosa of 5DN-treated mice, and the combined level of these metabolites in mouse colonic mucosa was 1.56-fold higher than that of 5DN. Cell culture studies revealed that 5DN and its colonic metabolites profoundly inhibited the growth of human colon cancer cells by inducing cell cycle arrest, triggering apoptosis and modulating key signaling proteins related to cell proliferation and apoptosis. Importantly, the colonic metabolites, especially M1, showed much stronger effects than those produced by 5DN itself. Overall, our results demonstrated that dietary 5DN significantly inhibited colitis-driven colon carcinogenesis in mice, and this chemopreventive effect was associated with its metabolites in the colon.
Collapse
Affiliation(s)
- Mingyue Song
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong, P.R. China and Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong, P.R. China and Department of Food Science, University of Massachusetts, Amherst, MA, USA.
| | - Yaqi Lan
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong, P.R. China and Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong, P.R. China
| | - Xian Wu
- Department of Food Science, University of Massachusetts, Amherst, MA, USA. and Department of Kinesiology and Health, Miami University, Oxford, OH, USA
| | - Yanhui Han
- Department of Food Science, University of Massachusetts, Amherst, MA, USA.
| | - Minqi Wang
- Department of Food Science, University of Massachusetts, Amherst, MA, USA.
| | - Jinkai Zheng
- Department of Food Science, University of Massachusetts, Amherst, MA, USA. and Institute of Agro-Products Processing Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, P.R. China
| | - Zhengze Li
- Department of Food Science, University of Massachusetts, Amherst, MA, USA.
| | - Fang Li
- Department of Food Science, University of Massachusetts, Amherst, MA, USA.
| | - Jiazhi Zhou
- Department of Food Science, University of Massachusetts, Amherst, MA, USA.
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong, P.R. China and Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong, P.R. China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong, P.R. China and Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong, P.R. China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
44
|
Ghram M, Bonnet-Magnaval F, Hotea DI, Doran B, Ly S, DesGroseillers L. Staufen1 is Essential for Cell-Cycle Transitions and Cell Proliferation Via the Control of E2F1 Expression. J Mol Biol 2020; 432:3881-3897. [DOI: 10.1016/j.jmb.2020.04.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 12/16/2022]
|
45
|
Samuelsen L, Hansen PE, Vang O. Derivatives of usnic acid cause cytostatic effect in Caco-2 cells. Nat Prod Res 2020; 35:4953-4959. [PMID: 32352327 DOI: 10.1080/14786419.2020.1756796] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Usnic acid has anti-cancer activity, however, low solubility and toxicity limit the potential. To investigate biological activity of usnic acid derivatives, enantiopure derivatives were synthesised by reacting usnic acid with ethylenediamine, which yielded one dimer product ((+)-1), and two tetra cyclic compounds ((+)-2 and (-)-2). The products were characterised with NMR, and evaluated in vitro in human colon cancer cell line Caco-2 by cell count, phase-contrast microscopy, MTT-assay, measurement of DNA content and cell cycle distribution. All compounds tested showed cytostatic effect in Caco-2 cells, but each compound had a distinct cellular effect. Compound (+)-1 showed anti-proliferative activity by increasing the percentage of cells in S-phase with 25% compared to the control. Compounds (+)-2 and (-)-2 induced paraptosis, but only compound (+)-2 modulated cell cycle distribution by accumulating cells in G2/M-phase by 47% and reduced DNA content by 60%. All compounds express interesting cellular and potential anti-proliferative activity.
Collapse
Affiliation(s)
- Lisa Samuelsen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Poul Erik Hansen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Ole Vang
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| |
Collapse
|
46
|
Chiang CY, Hsu KD, Lin YY, Hsieh CW, Liu JM, Lu TY, Cheng KC. The Antiproliferation Activity of Ganoderma formosanum Extracts on Prostate Cancer Cells. MYCOBIOLOGY 2020; 48:219-227. [PMID: 37970558 PMCID: PMC10635139 DOI: 10.1080/12298093.2020.1746064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/12/2020] [Accepted: 03/15/2020] [Indexed: 11/17/2023]
Abstract
Androgen-independent prostate cancer accounts for mortality in the world. In this study, various extracts of a medical fungus dubbed Ganoderma formosanum were screened for inhibition of DU145 cells, an androgen-independent prostate cancer cell line. Results demonstrated that both hexane (GF-EH) and butanol (GF-EB) fraction of G. formosanum ethanol extract inhibited DU145 cell viability in a dose-dependent manner. GF-EH induced cell-cycle arrest in G1 phase of DU145 cells via downregulation of cyclin E2 protein expression. In addition, GF-EB triggered extrinsic apoptosis of DU145 cells by activating caspase 3 gene expression resulting in programed cell death. Above all, both GF-EH and GF-EB show lower toxicity to normal human fibroblast cell line compared to DU145 cell, implying that they possess specific drug action on cancer cells. This study provides a molecular basis of G. formosanum extract as a potential ingredient for treatment of androgen-independent prostate cancer.
Collapse
Affiliation(s)
- Cheng-Yen Chiang
- Division of Urology, Department of Surgery, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan City, Taiwan
| | - Kai-Di Hsu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Yen-Yi Lin
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Chang-Wei Hsieh
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Jui-Ming Liu
- Division of Urology, Department of Surgery, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan City, Taiwan
| | - Tze-Ying Lu
- Department of Internal Medicine, Taipei Medical University–Shuang Ho Hospital, New Taipei City, Taiwan
| | - Kuan-Chen Cheng
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Food Science Technology, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China. Medical University, Taichung, Taiwan
| |
Collapse
|
47
|
Khan F, Singh VK, Saeed M, Kausar MA, Ansari IA. Carvacrol Induced Program Cell Death and Cell Cycle Arrest in Androgen-Independent Human Prostate Cancer Cells via Inhibition of Notch Signaling. Anticancer Agents Med Chem 2020; 19:1588-1608. [PMID: 31364516 DOI: 10.2174/1871520619666190731152942] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 05/09/2019] [Accepted: 07/01/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Several studies have revealed that abnormal activation of Notch signaling is closely related with the development and progression of prostate cancer. Although there are numerous therapeutic strategies, a more effective modality with least side effects is urgently required for the treatment of prostate cancer. Carvacrol is a monoterpenoid phenol and majorly present in the essential oils of Lamiaceae family plants. Many previous reports have shown various biological activities of carvacrol like antioxidant, antiinflammatory and anticancer properties. Recently, we have shown potent anticancer property of carvacrol against prostate cancer cell line DU145. In the current study, we report the chemopreventive and therapeutic potential of carvacrol against another prostate cancer cell line PC-3 with its detailed mechanism of action. METHODS To determine the effect of the carvacrol on prostate cancer cells, the cell viability was estimated by MTT assay and cell death was estimated by LDH release assay. The apoptotic assay was performed by DAPI staining and FITC-Annexin V assay. Reactive Oxygen Species (ROS) was estimated by DCFDA method. Cell cycle analysis was performed by flow cytometry. Gene expression analysis was performed by quantitative real time PCR. RESULTS Our results suggested that the carvacrol treatment significantly reduced the cell viability of PC-3 cells in a dose- and time-dependent manner. The antiproliferative action of carvacrol was correlated with apoptosis which was confirmed by nuclear condensation, FITC-Annexin V assay, modulation in expression of Bax, Bcl-2 and caspase activation. The mechanistic insight into carvacrol-induced apoptosis leads to finding of elevated level of Reactive Oxygen Species (ROS) and mitochondrial membrane potential disruption. Cell cycle analysis revealed that carvacrol prevented cell cycle in G0/G1 that was associated with decline in expression of cyclin D1 and Cyclin-Dependent Kinase 4 (CDK4) and augmented expression of CDK inhibitor p21. Having been said the role of hyperactivation of Notch signaling in prostate cancer, we also deciphered that carvacrol could inhibit Notch signaling in PC-3 cells via downregulation of Notch-1, and Jagged-1. CONCLUSION Thus, our previous and current findings have established the strong potential of carvacrol as a chemopreventive agent against androgen-independent human prostate cancer cells.
Collapse
Affiliation(s)
- Fahad Khan
- Department of Biosciences, Integral University, Dasauli, Kursi Road, Lucknow, 226026, India.,Noida Institute of Engineering and Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida, 201306, India
| | - Vipendra K Singh
- Environmental Carcinogenesis Laboratory, Food Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, Hail, Saudi Arabia
| | - Mohd A Kausar
- Department of Biochemistry, College of Medicine, University of Hail, Hail, Saudi Arabia
| | - Irfan A Ansari
- Department of Biosciences, Integral University, Dasauli, Kursi Road, Lucknow, 226026, India
| |
Collapse
|
48
|
Cell cycle synchronisation using thiazolidinediones affects cellular glucose metabolism and enhances the therapeutic effect of 2-deoxyglucose in colon cancer. Sci Rep 2020; 10:4713. [PMID: 32170185 PMCID: PMC7070066 DOI: 10.1038/s41598-020-61661-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/24/2020] [Indexed: 11/08/2022] Open
Abstract
The effect of cell cycle synchronisation on glucose metabolism in cancer cells is not known. We assessed how cell cycle synchronisation by thiazolidinediones (TZDs) can affect glucose uptake by cancer cells and investigated the anti-cancer effect of combination therapy with TZDs and 2-deoxy-glucose (2-DG) in colon cancer cells and in mouse xenograft models. Troglitazone (58.1 ± 2.0 vs 48.6 ± 1.3%, p = 0.002) or pioglitazone (82.9 ± 1.9 vs 61.6 ± 3.4%, p < 0.001) induced cell cycle arrest in SW480 cells at G1 phase. Western blot analysis showed the degradation of cyclin D1 and CDK4, and an increase in the expression levels of p21 and p27 after TZDs treatment. Withdrawal of troglitazone treatment induced significant increase in cellular 3H-DG uptake (141.5% ± 12.9% of controls) and membrane GLUT1 expression levels (146.3% of controls) by 24 h; 1 mM 2-DG treatment alone decreased cell survival by 5.8% as compared with the controls.; however, combination therapy enhanced the anti-tumour effects to 34.6% or 20.3% as compared with control cells. In vivo, each combination treatment group showed significant anti-tumour effects unlike the 2-DG alone group. Cell cycle synchronisation using TZDs induced cellular glucose uptake, which significantly enhanced the therapeutic effect of 2-DG in colon cancer.
Collapse
|
49
|
Pandey P, Bajpai P, Siddiqui MH, Sayyed U, Tiwari R, Shekh R, Mishra K, Kapoor VK. Elucidation of the Chemopreventive Role of Stigmasterol Against Jab1 in Gall Bladder Carcinoma. Endocr Metab Immune Disord Drug Targets 2020; 19:826-837. [PMID: 30727937 DOI: 10.2174/1871530319666190206124120] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/06/2018] [Accepted: 01/19/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Plant sterols have proven a potent anti-proliferative and apoptosis inducing agent against several carcinomas including breast and prostate cancers. Jab1 has been reported to be involved in the progression of numerous carcinomas. However, antiproliferative effects of sterols against Jab1 in gall bladder cancer have not been explored yet. OBJECTIVE In the current study, we elucidated the mechanism of action of stigmasterol regarding apoptosis induction mediated via downregulation of Jab1 protein in human gall bladder cancer cells. METHODS In our study, we performed MTT and Trypan blue assay to assess the effect of stigmasterol on cell proliferation. In addition, RT-PCR and western blotting were performed to identify the effect of stigmasterol on Jab1 and p27 expression in human gall bladder cancer cells. We further performed cell cycle, Caspase-3, Hoechst and FITC-Annexin V analysis, to confirm the apoptosis induction in stigmasterol treated human gall bladder cancer cells. RESULTS Our results clearly indicated that stigmasterol has up-regulated the p27 expression and down-regulated Jab1 gene. These modulations of genes might occur via mitochondrial apoptosis signaling pathway. Caspase-3 gets activated with the apoptotic induction. Increase in apoptotic cells and DNA were confirmed through annexin V staining, Hoechst staining, and cell cycle analysis. CONCLUSION Thus, these results strongly suggest that stigmasterol has the potential to be considered as an anticancerous therapeutic agent against Jab1 in gall bladder cancer.
Collapse
Affiliation(s)
- Pratibha Pandey
- Department of Bioengineering, Integral University, Lucknow, India.,Department of Biotechnology, Noida Institute of Engineering & Technology, Greater Noida, India
| | - Preeti Bajpai
- Department of Biosciences, Integral University, Lucknow, India
| | | | - Uzma Sayyed
- Department of Biosciences, Integral University, Lucknow, India
| | - Rohit Tiwari
- Department of Biosciences, Integral University, Lucknow, India
| | - Rafia Shekh
- Department of Biosciences, Integral University, Lucknow, India
| | - Kumudesh Mishra
- Department of Surgical Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - V K Kapoor
- Department of Surgical Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| |
Collapse
|
50
|
Reddy D, Ghosh P, Kumavath R. Strophanthidin Attenuates MAPK, PI3K/AKT/mTOR, and Wnt/β-Catenin Signaling Pathways in Human Cancers. Front Oncol 2020; 9:1469. [PMID: 32010609 PMCID: PMC6978703 DOI: 10.3389/fonc.2019.01469] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/09/2019] [Indexed: 12/16/2022] Open
Abstract
Lung cancer is the most prevalent in cancer-related deaths, while breast carcinoma is the second most dominant cancer in women, accounting for the most number of deaths worldwide. Cancers are heterogeneous diseases that consist of several subtypes based on the presence or absence of hormone receptors and human epidermal growth factor receptor 2. Several drugs have been developed targeting cancer biomarkers; nonetheless, their efficiency are not adequate due to the high reemergence rate of cancers and fundamental or acquired resistance toward such drugs, which leads to partial therapeutic possibilities. Recent studies on cardiac glycosides (CGs) positioned them as potent cytotoxic agents that target multiple pathways to initiate apoptosis and autophagic cell death in many cancers. In the present study, our aim is to identify the anticancer activity of a naturally available CG (strophanthidin) in human breast (MCF-7), lung (A549), and liver cancer (HepG2) cells. Our results demonstrate a dose-dependent cytotoxic effect of strophanthidin in MCF-7, A549, and HepG2 cells, which was further supported by DNA damage on drug treatment. Strophanthidin arrested the cell cycle at the G2/M phase; this effect was further validated by checking the inhibited expressions of checkpoint and cyclin-dependent kinases in strophanthidin-induced cells. Moreover, strophanthidin inhibited the expression of several key proteins such as MEK1, PI3K, AKT, mTOR, Gsk3α, and β-catenin from MAPK, PI3K/AKT/mTOR, and Wnt/β-catenin signaling. The current study adequately exhibits the role of strophanthidin in modulating the expression of various key proteins involved in cell cycle arrest, apoptosis, and autophagic cell death. Our in silico studies revealed that strophanthidin can interact with several key proteins from various pathways. Taken together, this study demonstrates the viability of strophanthidin as a promising anticancer agent, which may serve as a new anticancer drug.
Collapse
Affiliation(s)
- Dhanasekhar Reddy
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Kasaragod, India
| | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA, United States
| | - Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Kasaragod, India
| |
Collapse
|