1
|
Essid A, Elbini I, Ksiksi R, Harrab N, Moslah W, Jendoubi I, Doghri R, Zid MF, Luis J, Srairi-Abid N. Decavanadate Compound Displays In Vitro and In Vivo Antitumor Effect on Melanoma Models. Bioinorg Chem Appl 2025; 2025:6680022. [PMID: 39834888 PMCID: PMC11742080 DOI: 10.1155/bca/6680022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/29/2024] [Indexed: 01/22/2025] Open
Abstract
The efficacy of available treatments for melanoma is limited by side effects and the rapidly emerging resistance to treatment. In this context, the decavanadate compounds represent promising tools to design efficient therapeutic agents. In our study, we synthesized a dimagnesium disodium decavanadate icosahydrate compound (Mg2Na2V10O28·20H2O) and investigated its structure stability as well as its antimelanoma effects. Results showed that the Mg2Na2V10O28·20H2O compound is structured in a monoclinic system with the space group C2/c, stabilized by oxygen vertices, hydrogen bonds, and van der Waals interactions. Interestingly, we found that this newly synthesized compound reduced the viability of human (IGR39, IGR37, and SKMEL28) and murine (B16-F10) melanoma cells in a dose-dependent manner. The IC50 values ranged from 7.3 to 18 μM after 24 h and decreased to 1.4 μM after 72 h of treatment. Notably, this effect was more important than that of cisplatin (IC50 = 3 μM after 72 h of treatment), a chemotherapeutic agent, commonly used in the treatment of malignant melanoma. Furthermore, the cytotoxicity of the decavanadate compound was relatively weak on normal human keratinocytes (HaCaT), with a light effect (IC50 >> 70 μM) observed after 24 h of treatment. Thus, the Mg2Na2V10O28·20H2O compound displayed an advantage over cisplatin, which was reported to be much more aggressive to the keratinocyte cell line (IC50 = 23.9 μM). Moreover, it inhibited dose-dependently the adhesion of IGR39 cells to collagen (IC50 = 2.67 μM) and fibronectin, as well as their migration with an IC50 value of 2.23 μM. More interestingly, its in vivo administration to B16-F10 allografted mice, at the nontoxic dose of 50 μg (2.5 mg/kg), prevented and suppressed by 70% the tumor growth, compared to the nontreated mice. Moreover, this compound has also allowed a recovery against inflammation induced in mice by a mixture of DMBA and croton oil. Thus, all our results showed the potential of the Mg2Na2V10O28·20H2O compound to prevent and efficiently treat the growth and metastasis of melanoma.
Collapse
Affiliation(s)
- Amine Essid
- Institut Pasteur de Tunis, LR20IPT01 Biomolécules, Venins et Application Théranostiques (LBVAT), University of Tunis El Manar, Tunis 1002, Tunisia
| | - Ines Elbini
- Institut Pasteur de Tunis, LR20IPT01 Biomolécules, Venins et Application Théranostiques (LBVAT), University of Tunis El Manar, Tunis 1002, Tunisia
| | - Regaya Ksiksi
- Faculty of Sciences of Tunis, Laboratory of Materials, Crystal Chemistry and Applied Thermodynamics, University of Tunis El Manar, El Manar II, Tunis 2092, Tunisia
| | - Nardine Harrab
- Institut Pasteur de Tunis, LR20IPT01 Biomolécules, Venins et Application Théranostiques (LBVAT), University of Tunis El Manar, Tunis 1002, Tunisia
| | - Wassim Moslah
- Institut Pasteur de Tunis, LR20IPT01 Biomolécules, Venins et Application Théranostiques (LBVAT), University of Tunis El Manar, Tunis 1002, Tunisia
| | - Imen Jendoubi
- Faculty of Sciences of Tunis, Laboratory of Materials, Crystal Chemistry and Applied Thermodynamics, University of Tunis El Manar, El Manar II, Tunis 2092, Tunisia
| | - Raoudha Doghri
- Laboratoire de Médecine de Précision, Médecine Personnalisée et Investigation en Oncologie (LR21SP01), Service d'Anatomie Pathologique, Institut Salah Azaiez, Bab Saadoun, Tunis 1006, Tunisia
| | - Mohamed-Faouzi Zid
- Faculty of Sciences of Tunis, Laboratory of Materials, Crystal Chemistry and Applied Thermodynamics, University of Tunis El Manar, El Manar II, Tunis 2092, Tunisia
| | - José Luis
- Institut de Neurophysiopathologie, INP, Faculté des Sciences Médicales et Paramédicales, CNRS, Aix-Marseille Université, Marseille 13005, France
| | - Najet Srairi-Abid
- Institut Pasteur de Tunis, LR20IPT01 Biomolécules, Venins et Application Théranostiques (LBVAT), University of Tunis El Manar, Tunis 1002, Tunisia
| |
Collapse
|
2
|
Bano N, Parveen S, Saeed M, Siddiqui S, Abohassan M, Mir SS. Drug Repurposing of Selected Antibiotics: An Emerging Approach in Cancer Drug Discovery. ACS OMEGA 2024; 9:26762-26779. [PMID: 38947816 PMCID: PMC11209889 DOI: 10.1021/acsomega.4c00617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/19/2024] [Accepted: 05/29/2024] [Indexed: 07/02/2024]
Abstract
Drug repurposing is a method of investigating new therapeutic applications for previously approved medications. This repurposing approach to "old" medications is now highly efficient, simple to arrange, and cost-effective and poses little risk of failure in treating a variety of disorders, including cancer. Drug repurposing for cancer therapy is currently a key topic of study. It is a way of exploring recent therapeutic applications for already-existing drugs. Theoretically, the repurposing strategy has various advantages over the recognized challenges of creating new molecular entities, including being faster, safer, easier, and less expensive. In the real world, several medications have been repurposed, including aspirin, metformin, and chloroquine. However, doctors and scientists address numerous challenges when repurposing drugs, such as the fact that most drugs are not cost-effective and are resistant to bacteria. So the goal of this review is to gather information regarding repurposing pharmaceuticals to make them more cost-effective and harder for bacteria to resist. Cancer patients are more susceptible to bacterial infections. Due to their weak immune systems, antibiotics help protect them from a variety of infectious diseases. Although antibiotics are not immune boosters, they do benefit the defense system by killing bacteria and slowing the growth of cancer cells. Their use also increases the therapeutic efficacy and helps avoid recurrence. Of late, antibiotics have been repurposed as potent anticancer agents because of the evolutionary relationship between the prokaryotic genome and mitochondrial DNA of eukaryotes. Anticancer antibiotics that prevent cancer cells from growing by interfering with their DNA and blocking growth of promoters, which include anthracyclines, daunorubicin, epirubicin, mitoxantrone, doxorubicin, and idarubicin, are another type of FDA-approved antibiotics used to treat cancer. According to the endosymbiotic hypothesis, prokaryotes and eukaryotes are thought to have an evolutionary relationship. Hence, in this study, we are trying to explore antibiotics that are necessary for treating diseases, including cancer, helping people reduce deaths associated with various infections, and substantially extending people's life expectancy and quality of life.
Collapse
Affiliation(s)
- Nilofer Bano
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow 226026, India
- Department
of Bioengineering, Faculty of Engineering, Integral University, Kursi Road, Lucknow 226026, India
| | - Sana Parveen
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow 226026, India
- Department
of Biosciences, Faculty of Science, Integral
University, Kursi Road, Lucknow 226026, India
| | - Mohd Saeed
- Department
of Biology, College of Sciences, University
of Hail, P.O. Box 2240, Hail 55476, Saudi Arabia
| | - Samra Siddiqui
- Department
of Health Services Management, College of Public Health and Health
Informatics, University of Hail, Hail 55476, Saudi Arabia
| | - Mohammad Abohassan
- Department
of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
| | - Snober S. Mir
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow 226026, India
- Department
of Biosciences, Faculty of Science, Integral
University, Kursi Road, Lucknow 226026, India
| |
Collapse
|
3
|
Bano S, Alburquerque JQ, Roberts HJ, Pang S, Huang HC, Hasan T. Minocycline and photodynamic priming significantly improve chemotherapy efficacy in heterotypic spheroids of pancreatic ductal adenocarcinoma. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 255:112910. [PMID: 38663337 PMCID: PMC11088523 DOI: 10.1016/j.jphotobiol.2024.112910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/09/2024] [Accepted: 04/15/2024] [Indexed: 05/13/2024]
Abstract
The prognosis for patients with advanced-stage pancreatic ductal adenocarcinoma (PDAC) remains dismal. It is generally accepted that combination cancer therapies offer the most promise, such as Folforinox, despite their associated high toxicity. This study addresses the issue of chemoresistance by introducing a complementary dual priming approach to attenuate the DNA repair mechanism and to improve the efficacy of a type 1 topoisomerase (Top1) inhibitor. The result is a regimen that integrates drug-repurposing and nanotechnology using 3 clinically relevant FDA-approved agents (1) Top1 inhibitor (irinotecan) at subcytotoxic doses (2) benzoporphyrin derivative (BPD) as a photoactive molecule for photodynamic priming (PDP) to improve the delivery of irinotecan within the cancer cell and (3) minocycline priming (MNP) to modulate DNA repair enzyme Tdp1 (tyrosyl-DNA phosphodiesterase) activity. We demonstrate in heterotypic 3D cancer models that incorporate cancer cells and pancreatic cancer-associated fibroblasts that simultaneous targeting of Tdp1 and Top1 were significantly more effective by employing MNP and photoactivatable multi-inhibitor liposomes encapsulating BPD and irinotecan compared to monotherapies or a cocktail of dual or triple-agents. These data are encouraging and warrant further work in appropriate animal models to evolve improved therapeutic regimens.
Collapse
Affiliation(s)
- Shazia Bano
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, MA, USA
| | - Jose Quilez Alburquerque
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, MA, USA
| | - Harrison James Roberts
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, MA, USA
| | - Sumiao Pang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, MA, USA; Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, MA, USA.
| |
Collapse
|
4
|
Rezaei A, Moqadami A, Khalaj-Kondori M. Minocycline as a prospective therapeutic agent for cancer and non-cancer diseases: a scoping review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2835-2848. [PMID: 37991540 DOI: 10.1007/s00210-023-02839-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 11/05/2023] [Indexed: 11/23/2023]
Abstract
Minocycline is an FDA-approved secondary-generation tetracycline antibiotic. It is a synthetic antibiotic having many biological effects, such as antioxidant, anti-inflammatory, anti-cancer, and neuroprotective functions. This study discusses the pharmacological mechanisms of preventive and therapeutic effects of minocycline. Specifically, it provides a comprehensive overview of the molecular pathways by which minocycline acts on the different cancers, including ovarian, breast, glioma, colorectal, liver, pancreatic, lung, prostate, melanoma, head and neck, leukemia, and non-cancer diseases such as Alzheimer's disease, Parkinson, schizophrenia, multiple sclerosis, Huntington, polycystic ovary syndrome, and coronavirus disease 19. Minocycline may be a potential medication for these disorders due to its strong blood-brain barrier penetrance. It is also widely accepted as a specific medication, has a well-known side-effect characteristic, is reasonably priced, making it appropriate for continuous use in managing diseases, and has been demonstrated as an oral approach because it is effectively absorbed and accomplished almost all of the body's parts.
Collapse
Affiliation(s)
- Abedeh Rezaei
- Department of Animal Biology¸ Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Amin Moqadami
- Department of Animal Biology¸ Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology¸ Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
5
|
Rezaei A, Moqadami A, Khalaj-Kondori M, Feizi MAH. Minocycline induced apoptosis and suppressed expression of matrix metalloproteinases 2 and 9 in the breast cancer MCF-7 cells. Mol Biol Rep 2024; 51:463. [PMID: 38551800 DOI: 10.1007/s11033-024-09380-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 02/26/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND In women, breast cancer is the second most frequent type of cancer. Looking for new and effective cancer-specific therapies with little to no adverse effects on healthy cells is critical. OBJECTIVE Minocycline, a second-generation tetracycline, has shown anticancer effects by targeting multiple pathways in various cancers. This study aimed to determine minocycline effects on the cell proliferation, apoptosis, and invasion of the human MCF-7 cells. METHODS MTT assay was used to evaluate the cytotoxicity of minocycline on the cells. Flow cytometry was performed to investigate the induction of apoptosis and the cell cycle progression. The expression levels of apoptotic and migration proteins and genes were assessed by western blotting and qRT-PCR. The scratch test was performed to evaluate the anti-migration effect of the drug. RESULTS The results indicated that the IC50 value of minocycline for MCF-7 cells was 36.10 µM. Minocycline treatment caused sub-G1 cell accumulation, indicating a significant apoptotic effect on the MCF-7 cells. Annexin-V/PI staining revealed a significant rise in early and late apoptotic cell percentages. Minocycline up-regulated Bax and Caspase-3 expression and down-regulated Bcl-2 and Pro-Cas3. The scratch test revealed significant anti-migration effects for minocycline. Furthermore, it caused down-regulation of MMP-2 and MMP-9 in a concentration-dependent method. CONCLUSION These findings further confirmed the anticancer effect of minocycline and highlighted that minocycline maybe considered as potential therapeutic agent for breast cancer treatment.
Collapse
Affiliation(s)
- Abedeh Rezaei
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Amin Moqadami
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | | |
Collapse
|
6
|
Paneth A, Kaproń B, Plech T, Paduch R, Trotsko N, Paneth P. Combined In Silico and In Vitro Analyses to Assess the Anticancer Potential of Thiazolidinedione-Thiosemicarbazone Hybrid Molecules. Int J Mol Sci 2023; 24:17521. [PMID: 38139350 PMCID: PMC10743653 DOI: 10.3390/ijms242417521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/09/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
The number of people affected by cancer and antibiotic-resistant bacterial infections has increased, such that both diseases are already seen as current and future leading causes of death globally. To address this issue, based on a combined in silico and in vitro approach, we explored the anticancer potential of known antibacterials with a thiazolidinedione-thiosemicarbazone (TZD-TSC) core structure. A cytotoxicity assessment showed encouraging results for compounds 2-4, with IC50 values against T98G and HepG2 cells in the low micromolar range. TZD-TSC 3 proved to be most toxic to cancer cell lines, with IC50 values of 2.97 ± 0.39 µM against human hepatoma HepG2 cells and IC50 values of 28.34 ± 2.21 µM against human glioblastoma T98G cells. Additionally, compound 3 induced apoptosis and showed no specific hemolytic activity. Furthermore, treatment using 3 on cancer cell lines alters these cells' morphology and further suppresses migratory activity. Molecular docking, in turn, suggests that 3 would have the capacity to simultaneously target HDACs and PPARγ, by the activation of PPARγ and the inhibition of both HDAC4 and HDAC8. Thus, the promising preliminary results obtained with TZD-TSC 3 represent an encouraging starting point for the rational design of novel chemotherapeutics with dual antibacterial and anticancer activities.
Collapse
Affiliation(s)
- Agata Paneth
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, 20-059 Lublin, Poland;
| | - Barbara Kaproń
- Department of Clinical Genetics, Faculty of Medicine, Medical University of Lublin, 20-080 Lublin, Poland
| | - Tomasz Plech
- Department of Pharmacology, Faculty of Health Sciences, Medical University of Lublin, 20-080 Lublin, Poland;
| | - Roman Paduch
- Department of Virology and Immunology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, 20-033 Lublin, Poland;
| | - Nazar Trotsko
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, 20-059 Lublin, Poland;
| | - Piotr Paneth
- Institute of Applied Radiation Chemistry, Faculty of Chemistry, Lodz University of Technology, 90-924 Lodz, Poland
| |
Collapse
|
7
|
Li J, Qin Y, Zhao C, Zhang Z, Zhou Z. Tetracycline antibiotics: Potential anticancer drugs. Eur J Pharmacol 2023; 956:175949. [PMID: 37541377 DOI: 10.1016/j.ejphar.2023.175949] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/22/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
In recent years, research on tetracycline antibiotics has gradually shifted from their antibacterial effects to anticancer effects. Doxycycline, minocycline, and tigecycline as the US Food and Drug Administration (FDA) approved tetracycline antibiotics have been the main subjects of studies. Evidence indicated that they have anticancer properties and are able to control cancer progression through different mechanisms, such as anti-proliferation, anti-metastasis, and promotion of autophagy or apoptosis. In addition, studies have shown that these three tetracycline antibiotics can be utilized in conjunction with chemotherapeutic and targeted drugs to inhibit cancer progression and improve the quality of patient survival. Therefore, doxycycline, minocycline, and tigecycline are taken as examples in this work. Their mechanisms of action in different cancers and related combination therapies are introduced. Their current roles in alleviating the suffering of patients undergoing chemotherapy when used as adjuvant drugs in clinical treatment are also described. Finally, the research gaps and potential research directions at this stage are briefly summarized.
Collapse
Affiliation(s)
- Jiayu Li
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yuan Qin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China; College of Pharmacy, Nankai University, China
| | - Chenhao Zhao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Zhi Zhang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Zhiruo Zhou
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou, China.
| |
Collapse
|
8
|
Rok J, Kowalska J, Rzepka Z, Stencel D, Skorek A, Banach K, Wrześniok D. The Assessment of Anti-Melanoma Potential of Tigecycline-Cellular and Molecular Studies of Cell Proliferation, Apoptosis and Autophagy on Amelanotic and Melanotic Melanoma Cells. Cells 2023; 12:1564. [PMID: 37371034 DOI: 10.3390/cells12121564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
High mortality, aggressiveness, and the relatively low effectiveness of therapy make melanoma the most dangerous of skin cancers. Previously published studies presented the promising therapeutic potential of minocycline, doxycycline, and chlortetracycline on melanoma cells. This study aimed to assess the cytotoxicity of tigecycline, a third-generation tetracycline, on melanotic (COLO 829) and amelanotic (A375) melanoma cell lines. The obtained results showed that tigecycline, proportionally to the concentration and incubation time, efficiently inhibited proliferation of both types of melanoma cells. The effect was accompanied by the dysregulation of the cell cycle, the depolarization of the mitochondrial membrane, and a decrease in the reduced thiols and the levels of MITF and p44/42 MAPK. However, the ability to induce apoptosis was only found in COLO 829 melanoma cells. A375 cells appeared to be more resistant to the treatment with tigecycline. The drug did not induce apoptosis but caused an increase in LC3A/B protein levels-an autophagy marker. The observed differences in drug action on the tested cell lines also involved an increase in p21 and p16 protein levels in melanotic melanoma, which was related to cell cycle arrest in the G1/G0 phase. The greater sensitivity of melanotic melanoma cells to the action of tigecycline suggests the possibility of considering the use of the drug in targeted therapy.
Collapse
Affiliation(s)
- Jakub Rok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| | - Justyna Kowalska
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| | - Zuzanna Rzepka
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| | - Dominika Stencel
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| | - Anna Skorek
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| | - Klaudia Banach
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| | - Dorota Wrześniok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| |
Collapse
|
9
|
The Assessment of the Phototoxic Action of Chlortetracycline and Doxycycline as a Potential Treatment of Melanotic Melanoma-Biochemical and Molecular Studies on COLO 829 and G-361 Cell Lines. Int J Mol Sci 2023; 24:ijms24032353. [PMID: 36768675 PMCID: PMC9917077 DOI: 10.3390/ijms24032353] [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: 12/14/2022] [Revised: 01/22/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023] Open
Abstract
Melanoma is still one of the most dangerous cancers. New methods of treatment are sought due to its high aggressiveness and the relatively low effectiveness of therapies. Tetracyclines are drugs exhibiting anticancer activity. Previous studies have also shown their activity against melanoma cells. The possibility of tetracycline accumulation in pigmented tissues and the increase in their toxicity under the influence of UVA radiation creates the possibility of developing a new anti-melanoma therapy. This study aimed to analyze the phototoxic effect of doxycycline and chlortetracycline on melanotic melanoma cells COLO 829 and G-361. The results indicated that tetracycline-induced phototoxicity significantly decreased the number of live cells by cell cycle arrest as well as a decrease in cell viability. The simultaneous exposure of cells to drugs and UVA caused the depolarization of mitochondria as well as inducing oxidative stress and apoptosis. It was found that the combined treatment activated initiator and effector caspases, caused DNA fragmentation and elevated p53 level. Finally, it was concluded that doxycycline demonstrated a stronger cytotoxic and phototoxic effect. UVA irradiation of melanoma cells treated with doxycycline and chlortetracycline allows for the reduction of therapeutic drug concentrations and increases the effectiveness of tested tetracyclines.
Collapse
|
10
|
Rzepka Z, Rok J, Kowalska J, Banach K, Wrześniok D. Cobalamin Deficiency May Induce Astrosenescence-An In Vitro Study. Cells 2022; 11:3408. [PMID: 36359805 PMCID: PMC9655094 DOI: 10.3390/cells11213408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/19/2022] [Accepted: 10/26/2022] [Indexed: 11/30/2022] Open
Abstract
Cobalamin (vitamin B12) deficiency is one of the major factors causing degenerative changes in the nervous system and, thus, various neurological and psychiatric symptoms. The underlying cellular mechanism of this phenomenon is not yet fully understood. An accumulation of senescent astrocytes has been shown to contribute to a wide range of pathologies of the nervous system, including neurodegenerative disorders. This study aimed to investigate whether cobalamin deficiency triggers astrosenescence. After inducing cobalamin deficiency in normal human astrocytes in vitro, we examined biomarkers of cellular senescence: SA-β-gal, p16INK4A, and p21Waf1/Cip1 and performed cell nuclei measurements. The obtained results may contribute to an increase in the knowledge of the cellular effects of cobalamin deficiency in the context of astrocytes. In addition, the presented data suggest a potential causative agent of astrosenescence that has not been proven to date.
Collapse
Affiliation(s)
- Zuzanna Rzepka
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| | | | | | | | | |
Collapse
|
11
|
New Betulin Derivatives with Nitrogen Heterocyclic Moiety-Synthesis and Anticancer Activity In Vitro. Biomolecules 2022; 12:biom12101540. [PMID: 36291749 PMCID: PMC9599051 DOI: 10.3390/biom12101540] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/15/2022] [Accepted: 10/19/2022] [Indexed: 12/01/2022] Open
Abstract
As part of the search for new medicinal substances with potential application in oncology, the synthesis of new compounds combining the betulin molecule and the indole system was carried out. The structure of the ester derivatives obtained in the Steglich reaction was confirmed by spectroscopic methods (1H and 13C NMR, HR-MS). The obtained new 3-indolyl betulin derivatives were evaluated for anticancer activity against several human cancer cell lines (melanomas, breast cancers, colorectal adenocarcinomas, lung cancer) as well as normal human fibroblasts. The significant reduction in MCF-7 cells viability for 28-hydroxy-(lup-20(29)-ene)-3-yl 2-(1H-indol-3-yl)acetate was observed at a concentration of 10 µg/mL (17 µM). In addition, cytometric analysis showed that this compound strongly reduces the proliferation rate of breast cancer cells. For this, the derivative showing the promising cytotoxic effect on MCF-7 breast cancer cells, the pharmacokinetic profile prediction was performed using in silico methods. Based on the results obtained in the study, it can be concluded that indole-functionalized triterpene EB367 is a promising starting point for further research in the field of breast cancer therapy or the synthesis of new derivatives.
Collapse
|
12
|
Kuruburu MG, Bovilla VR, Leihang Z, Madhunapantula SV. Phytochemical-rich Fractions from Foxtail Millet (Setaria italica (L.) P.Beauv) Seeds Exhibited Antioxidant Activity and Reduced the Viability of Breast Cancer Cells In Vitro by Inducing DNA Fragmentation and Promoting Cell Cycle Arrest. Anticancer Agents Med Chem 2022; 22:2477-2493. [PMID: 35168527 DOI: 10.2174/1871520622666220215122141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/02/2021] [Accepted: 12/20/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The demand for millet-based diets has increased significantly in recent years due to their beneficial effects on human health. Foxtail Millet (Setaria italica (L.) P.Beauv, previously known as Panicum italicum L., referred as FTM in this manuscript) seeds have been not only used as astringent and diuretic agents, but they are also used to treat dyspepsia and rheumatism. Recent studies have shown that solvent extracts from FTM seeds exhibited antioxidant and antiinflammatory activities. However, the nature and antiproliferative potential of phytochemical constituents of solvent extracts are not much explored. OBJECTIVES Major objectives of this study are to generate and characterize the phytochemical-rich fractions from Foxtail millet seeds, test the antioxidant activity and antiproliferative potential against cell lines representing carcinomas of the breast, and determine the mechanism(s) of cell growth inhibition. METHODS Phytochemical-rich fractions were generated by extracting the seeds using 70% ethanol (FTM-FP) and 10% alkali (FTM-BP). Antioxidant potential was determined by ferric reducing antioxidant power (FRAP) assay and DPPH radical scavenging activity assays. The antiproliferative potential was determined using sulforhodamine-B assay. The impact on cell cycle and DNA fragmentation was analyzed by staining the cells with DAPI followed by analyzing the stained cells using NC-3000. RESULTS Analysis of the results showed the presence of phenolics and flavonoids in the FTM-FP and FTM-BP fractions. Both fractions exhibited antiproliferative potential against breast cancer cell lines. Mechanistically, both fractions induced G2/M cell cycle arrest and increased the fragmentation of DNA, which lead to the accumulation of cells in the Sub-G1 phase. CONCLUSION In summary, results of this study demonstrated the potential of foxtail millet phytochemical fractions for retarding the proliferative potential of breast cancer cells.
Collapse
Affiliation(s)
- Mahadevaswamy G Kuruburu
- Center of Excellence in Molecular Biology and Regenerative Medicine (A DST-FIST Supported Center), Department of Biochemistry (A DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru - 570015, Karnataka, India
| | - Venugopal R Bovilla
- Center of Excellence in Molecular Biology and Regenerative Medicine (A DST-FIST Supported Center), Department of Biochemistry (A DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru - 570015, Karnataka, India
- Public Health Research Institute of India (PHRII), Yadavagiri, Mysuru - 570020, Karnataka, India
| | - Zonunsiami Leihang
- Center of Excellence in Molecular Biology and Regenerative Medicine (A DST-FIST Supported Center), Department of Biochemistry (A DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru - 570015, Karnataka, India
| | - SubbaRao V Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (A DST-FIST Supported Center), Department of Biochemistry (A DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru - 570015, Karnataka, India
- Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru - 570015, Karnataka, India
| |
Collapse
|
13
|
Rok J, Rzepka Z, Kowalska J, Banach K, Beberok A, Wrześniok D. The Anticancer Potential of Doxycycline and Minocycline-A Comparative Study on Amelanotic Melanoma Cell Lines. Int J Mol Sci 2022; 23:ijms23020831. [PMID: 35055021 PMCID: PMC8775630 DOI: 10.3390/ijms23020831] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/02/2022] [Accepted: 01/11/2022] [Indexed: 12/04/2022] Open
Abstract
Malignant melanoma is still a serious medical problem. Relatively high mortality, a still-growing number of newly diagnosed cases, and insufficiently effective methods of therapy necessitate melanoma research. Tetracyclines are compounds with pleiotropic pharmacological properties. Previously published studies on melanotic melanoma cells ascertained that minocycline and doxycycline exerted an anti-melanoma effect. The purpose of the study was to assess the anti-melanoma potential and mechanisms of action of minocycline and doxycycline using A375 and C32 human amelanotic melanoma cell lines. The obtained results indicate that the tested drugs inhibited proliferation, decreased cell viability, and induced apoptosis in amelanotic melanoma cells. The treatment caused changes in the cell cycle profile and decreased the intracellular level of reduced thiols and mitochondrial membrane potential. The exposure of A375 and C32 cells to minocycline and doxycycline triggered the release of cytochrome c and activated initiator and effector caspases. The anti-melanoma effect of analyzed drugs appeared to be related to the up-regulation of ERK1/2 and MITF. Moreover, it was noticed that minocycline and doxycycline increased the level of LC3A/B, an autophagy marker, in A375 cells. In summary, the study showed the pleiotropic anti-cancer action of minocycline and doxycycline against amelanotic melanoma cells. Considering all results, it could be concluded that doxycycline was a more potent drug than minocycline.
Collapse
Affiliation(s)
- Jakub Rok
- Correspondence: ; Tel.: +48-32-364-15-47
| | | | | | | | | | | |
Collapse
|
14
|
Wang YC, Ku WC, Liu CY, Cheng YC, Chien CC, Chang KW, Huang CJ. Supplementation of Probiotic Butyricicoccus pullicaecorum Mediates Anticancer Effect on Bladder Urothelial Cells by Regulating Butyrate-Responsive Molecular Signatures. Diagnostics (Basel) 2021; 11:diagnostics11122270. [PMID: 34943506 PMCID: PMC8700285 DOI: 10.3390/diagnostics11122270] [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: 09/28/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 11/30/2022] Open
Abstract
In bladder cancer, urothelial carcinoma is the most common histologic subtype, accounting for more than 90% of cases. Pathogenic effects due to the dysbiosis of gut microbiota are localized not only in the colon, but also in regulating bladder cancer distally. Butyrate, a short-chain fatty acid produced by gut microbial metabolism, is mainly studied in colon diseases. Therefore, the resolution of the anti-cancer effects of butyrate-producing microbes on bladder urothelial cells and knowledge of the butyrate-responsive molecules must have clinical significance. Here, we demonstrate a correlation between urothelial cancer of the bladder and Butyricicoccus pullicaecorum. This butyrate-producing microbe or their metabolite, butyrate, mediated anti-cancer effects on bladder urothelial cells by regulating cell cycle, cell growth, apoptosis, and gene expression. For example, a tumor suppressor against urothelial cancer of the bladder, bladder cancer-associated protein, was induced in butyrate-treated HT1376 cells, a human urinary bladder cancer cell line. In conclusion, urothelial cancer of the bladder is a significant health problem. To improve the health of bladder urothelial cells, supplementation of B. pullicaecorum may be necessary and can further regulate butyrate-responsive molecular signatures.
Collapse
Affiliation(s)
- Yen-Chieh Wang
- Department of Urology, Cathay General Hospital, Taipei 106438, Taiwan;
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 242062, Taiwan; (W.-C.K.); (C.-Y.L.); (Y.-C.C.); (C.-C.C.)
| | - Wei-Chi Ku
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 242062, Taiwan; (W.-C.K.); (C.-Y.L.); (Y.-C.C.); (C.-C.C.)
| | - Chih-Yi Liu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 242062, Taiwan; (W.-C.K.); (C.-Y.L.); (Y.-C.C.); (C.-C.C.)
- Department of Pathology, Sijhih Cathay General Hospital, New Taipei 221037, Taiwan
| | - Yu-Che Cheng
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 242062, Taiwan; (W.-C.K.); (C.-Y.L.); (Y.-C.C.); (C.-C.C.)
- Department of Medical Research, Cathay General Hospital, Taipei 106438, Taiwan
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan 320317, Taiwan
| | - Chih-Cheng Chien
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 242062, Taiwan; (W.-C.K.); (C.-Y.L.); (Y.-C.C.); (C.-C.C.)
- Department of Anesthesiology, Cathay General Hospital, Taipei 106438, Taiwan
| | - Kang-Wei Chang
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 110301, Taiwan;
- Laboratory Animal Center, Taipei Medical University, Taipei 110301, Taiwan
| | - Chi-Jung Huang
- Department of Medical Research, Cathay General Hospital, Taipei 106438, Taiwan
- Department of Biochemistry, National Defense Medical Center, Taipei 114201, Taiwan
- Correspondence:
| |
Collapse
|
15
|
Chikkegowda P, Pookunoth BC, Bovilla VR, Veeresh PM, Leihang Z, Thippeswamy T, Padukudru MA, Hathur B, Kanchugarakoppal RS, Madhunapantula SV. Design, Synthesis, Characterization, and Crystal Structure Studies of Nrf2 Modulators for Inhibiting Cancer Cell Growth In Vitro and In Vivo. ACS OMEGA 2021; 6:10054-10071. [PMID: 34056161 PMCID: PMC8153663 DOI: 10.1021/acsomega.0c06345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/24/2021] [Indexed: 05/03/2023]
Abstract
Nrf2 is one of the important therapeutic targets studied extensively in several cancers including the carcinomas of the colon and rectum. However, to date, not many Nrf2 inhibitors showed promising results for retarding the growth of colorectal cancers (CRCs). Therefore, in this study, first, we have demonstrated the therapeutic effect of siRNA-mediated downmodulation of Nrf2 on the proliferation rate of CRC cell lines. Next, we have designed, synthesized, characterized, and determined the crystal structures for a series of tetrahydrocarbazoles (THCs) and assessed their potential to modulate the activity of Nrf2 target gene NAD(P)H:quinone oxidoreductase (NQO1) activity by treating colorectal carcinoma cell line HCT-116. Later, the cytotoxic potential of compounds was assessed against cell lines expressing varying amounts of Nrf2, viz., breast cancer cell lines MDA-MB-231 and T47D (low functionally active Nrf2), HCT-116 (moderately active Nrf2), and lung cancer cell line A549 (highly active Nrf2), and the lead compound 5b was tested for its effect on cell cycle progression in vitro and for retarding the growth of Ehrlich ascites carcinomas (EACs) in mice. Data from our study demonstrated that among various compounds 5b exhibited better therapeutic index and retarded the growth of EAC cells in mice. Therefore, compound 5b is recommended for further development to target cancers.
Collapse
Affiliation(s)
- Prathima Chikkegowda
- Department
of Pharmacology, JSS Medical College, JSS
Academy of Higher Education & Research, Mysore 570015, Karnataka, India
| | - Baburajeev C. Pookunoth
- Laboratory
of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Mysore 570005, Karnataka, India
| | - Venugopal R. Bovilla
- Department
of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
- Center
of Excellence in Molecular Biology and Regenerative Medicine (CEMR,
DST-FIST Supported Center), JSS Medical College, JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
| | - Prashanthkumar M. Veeresh
- Department
of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
- Center
of Excellence in Molecular Biology and Regenerative Medicine (CEMR,
DST-FIST Supported Center), JSS Medical College, JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
| | - Zonunsiami Leihang
- Department
of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
- Center
of Excellence in Molecular Biology and Regenerative Medicine (CEMR,
DST-FIST Supported Center), JSS Medical College, JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
| | - Thippeswamy Thippeswamy
- Department
of General Medicine, JSS Medical College and Hospital, JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
| | - Mahesh A. Padukudru
- Department
of Respiratory Medicine, JSS Medical College, and Hospital, JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
| | - Basavanagowdappa Hathur
- Center
of Excellence in Molecular Biology and Regenerative Medicine (CEMR,
DST-FIST Supported Center), JSS Medical College, JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
- Department
of General Medicine, JSS Medical College and Hospital, JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
- Faculty
of Medicine, JSS Medical College and Hospital, JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
- JSS
Medical College and Hospital, JSS Academy
of Higher Education & Research, Mysore 570015, Karnataka, India
- Special
Interest Group in Patient Care Management, JSS Medical College and
Hospital, JSS Academy of Higher Education
& Research, Mysore 570015, Karnataka, India
| | | | - SubbaRao V. Madhunapantula
- Department
of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
- Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
- . Mobile: +91-810-527-8621
| |
Collapse
|
16
|
The role of UVA radiation in ketoprofen-mediated BRAF-mutant amelanotic melanoma cells death - A study at the cellular and molecular level. Toxicol In Vitro 2021; 72:105108. [PMID: 33545343 DOI: 10.1016/j.tiv.2021.105108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/08/2021] [Accepted: 02/01/2021] [Indexed: 12/27/2022]
Abstract
Malignant melanoma is the cause of 80% of deaths in skin cancer patients. Treatment of melanoma in the 4th stage of clinical advancement, in which inoperable metastasis occur, does not provide sufficient effects. Ketoprofen has phototoxic properties and it can be used as a new treatment option for skin cancers as a part of photochemotherapy. The present study was designed to investigate whether ketoprofen in combination with UVA induces cytotoxic, anti-proliferative and pro-apoptotic effects on melanoma cells. It was stated that co-treatment with 1.0 mM ketoprofen and UVA irradiation disturbed homeostasis of C32 melanoma cells by lowering its vitality (decrease of GSH level). Contrary to C32 cells, melanocytes showed low sensitivity to ketoprofen and UVA radiation, pointing selectivity in the mode of action towards melanoma cells. Co-treatment with ketoprofen and UVA irradiation has cytotoxic and anti-proliferative and pro-apoptotic effect on C32. The co-treatment triggered the DNA fragmentation and changed the cell cycle in C32 cells. In conclusion, it could be stated that local application of ketoprofen in combination with UVA irradiation may be used to support the treatment of melanoma and creates the possibility of reducing the risk of cancer recurrence and metastasis.
Collapse
|