1
|
Xu B, Huang Y, Yu D, Chen Y. Advancements of ROS-based biomaterials for sensorineural hearing loss therapy. Biomaterials 2025; 316:123026. [PMID: 39705924 DOI: 10.1016/j.biomaterials.2024.123026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/28/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Sensorineural hearing loss (SNHL) represents a substantial global health challenge, primarily driven by oxidative stress-induced damage within the auditory system. Excessive reactive oxygen species (ROS) play a pivotal role in this pathological process, leading to cellular damage and apoptosis of cochlear hair cells, culminating in irreversible hearing impairment. Recent advancements have introduced ROS-scavenging biomaterials as innovative, multifunctional platforms capable of mitigating oxidative stress. This comprehensive review systematically explores the mechanisms of ROS-mediated oxidative stress in SNHL, emphasizing etiological factors such as aging, acoustic trauma, and ototoxic medication exposure. Furthermore, it examines the therapeutic potential of ROS-scavenging biomaterials, positioning them as promising nanomedicines for targeted antioxidant intervention. By critically assessing recent advances in biomaterial design and functionality, this review thoroughly evaluates their translational potential for clinical applications. It also addresses the challenges and limitations of ROS-neutralizing strategies, while highlighting the transformative potential of these biomaterials in developing novel SNHL treatment modalities. This review advocates for continued research and development to integrate ROS-scavenging biomaterials into future clinical practice, aiming to address the unmet needs in SNHL management and potentially revolutionize the treatment landscape for this pervasive health issue.
Collapse
Affiliation(s)
- Baoying Xu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yuqi Huang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Dehong Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China; Shanghai Institute of Materdicine, Shanghai, 200012, China.
| |
Collapse
|
2
|
Pathak N, Negi AS. Plant based steroidal and triterpenoid sapogenins: Chemistry on diosgenin and biological aspects. Eur J Med Chem 2024; 279:116915. [PMID: 39366126 DOI: 10.1016/j.ejmech.2024.116915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/15/2024] [Accepted: 09/25/2024] [Indexed: 10/06/2024]
Abstract
Plants are rich in steroidal and triterpenoid saponins. Diosgenin is an important sapogenin obtained from various steroidal saponins and specially from dioscin. It possesses diverse pharmacological activities as it is capable of modulating various endogenous pathways. Diosgenin is the molecule of choice for the industrial synthesis of the steroid based clinical drugs namely progesterone, testosterone, dexamethasone, dehydroepiandrosterone, vitamin D3, steroidal contraceptive pills, norethindrone, norgestrel etc. Diosgenin has been a molecule of discussion due to its high demand in industry as well as for future research applications. Present review describes its chemistry and detailed pharmacological profile.
Collapse
Affiliation(s)
- Nandini Pathak
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants (CSIR-CIMAP, P.O. CIMAP, Kukrail Picnic Spot Road, Lucknow, 226015, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India
| | - Arvind S Negi
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants (CSIR-CIMAP, P.O. CIMAP, Kukrail Picnic Spot Road, Lucknow, 226015, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India.
| |
Collapse
|
3
|
Makhal PN, Sood A, Shaikh AS, Dayare LN, Khatri DK, Rao Kaki V. Development of trisubstituted thiophene-3-arboxamide selenide derivatives as novel EGFR kinase inhibitors with cytotoxic activity. RSC Med Chem 2023; 14:2677-2698. [PMID: 38107169 PMCID: PMC10718591 DOI: 10.1039/d3md00403a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 10/06/2023] [Indexed: 12/19/2023] Open
Abstract
Overexpression of EGFR is one of the eminent oncogenic drivers detected in the development of several human cancers. The increasing incidences of mutation-based resistance in the tyrosine kinase domain call upon the need for the development of a newer class of small-molecule TK inhibitors. Accordingly, a new series of symmetrical trisubstituted thiophene-3-carboxamide selenide derivatives was developed via the hybridization of complementary pharmacophores. Most of the compounds showed a modest to excellent antiproliferative action at 20 μM concentration. The utmost antiproliferative activity was portrayed by compound 16e on the selected cancer cell lines with IC50 < 9 μM, the lowest being 3.20 ± 0.12 μM in the HCT116 cell line. Further, it also displayed an impressive EGFR kinase inhibition with an IC50 value of 94.44 ± 2.22 nM concentration. As a corollary of the reported EGFR inhibition, the nature, energy, and stability of the binding interactions were contemplated via in silico studies.
Collapse
Affiliation(s)
- Priyanka N Makhal
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad-500037 India
| | - Anika Sood
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad-500037 India
| | - Arbaz Sujat Shaikh
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad-500037 India
| | - Lahu N Dayare
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad-500037 India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad-500037 India
| | - Venkata Rao Kaki
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad-500037 India
| |
Collapse
|
4
|
Jastrzebska I, Wawrusiewicz-Kurylonek N, Grześ PA, Ratkiewicz A, Grabowska E, Czerniecka M, Czyżewska U, Tylicki A. New Steroidal Selenides as Proapoptotic Factors. Molecules 2023; 28:7528. [PMID: 38005248 PMCID: PMC10673341 DOI: 10.3390/molecules28227528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/20/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Cytostatic and pro-apoptotic effects of selenium steroid derivatives against HeLa cells were determined. The highest cytostatic activity was shown by derivative 4 (GI50 25.0 µM, almost complete growth inhibition after three days of culture, and over 97% of apoptotic and dead cells at 200 µM). The results of our study (cell number measurements, apoptosis profile, relative expression of apoptosis-related APAF1, BID, and mevalonate pathway-involved HMGCR, SQLE, CYP51A1, and PDHB genes, and computational chemistry data) support the hypothesis that tested selenosteroids induce the extrinsic pathway of apoptosis by affecting the cell membrane as cholesterol antimetabolites. An additional mechanism of action is possible through a direct action of derivative 4 to inhibit PDHB expression in a way similar to steroid hormones.
Collapse
Affiliation(s)
- Izabella Jastrzebska
- Faculty of Chemistry, University of Białystok, Ciołkowskiego 1K, 15-245 Białystok, Poland
| | | | - Paweł A Grześ
- Faculty of Chemistry, University of Białystok, Ciołkowskiego 1K, 15-245 Białystok, Poland
| | - Artur Ratkiewicz
- Faculty of Chemistry, University of Białystok, Ciołkowskiego 1K, 15-245 Białystok, Poland
| | - Ewa Grabowska
- Doctoral School of Exact and Natural Sciences, University of Bialystok, K. Ciolkowskiego 1K, 15-245 Bialystok, Poland
| | - Magdalena Czerniecka
- Faculty of Biology, University of Białystok, Ciołkowskiego 1J, 15-245 Białystok, Poland
| | - Urszula Czyżewska
- Faculty of Biology, University of Białystok, Ciołkowskiego 1J, 15-245 Białystok, Poland
| | - Adam Tylicki
- Faculty of Biology, University of Białystok, Ciołkowskiego 1J, 15-245 Białystok, Poland
| |
Collapse
|
5
|
Roncero AM, Tobal IE, Moro RF, Diez D, Marcos IS. Halimanes and cancer: ent-halimic acid as a starting material for the synthesis of antitumor drugs. Front Chem 2023; 11:1225355. [PMID: 37674527 PMCID: PMC10477373 DOI: 10.3389/fchem.2023.1225355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/12/2023] [Indexed: 09/08/2023] Open
Abstract
The development of new anti-cancer agents is an urgent necessity nowadays, as it is one of the major causes of mortality worldwide. Many drugs currently used are derived from natural products. Halimanes are a class of bicyclic diterpenoids present in various plants and microorganisms. Many of them exhibit biological activities such as antitumor, antimicrobial, or anti-inflammatory. Among them, ent-halimic acid is an easily accessible compound, in large quantities, from the ethyl acetate extract of the plant Halimium viscosum, and it has been used as a starting material in a number of bioactive molecules. In this work, we review all the natural halimanes with antitumor and related activities until date as well as the synthesis of antitumor compounds using ent-halimic acid as a starting material.
Collapse
Affiliation(s)
| | | | | | | | - Isidro S. Marcos
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad de Salamanca, Salamanca, Spain
| |
Collapse
|
6
|
Astrain-Redin N, Raza A, Encío I, Sharma AK, Plano D, Sanmartín C. Novel Acylselenourea Derivatives: Dual Molecules with Anticancer and Radical Scavenging Activity. Antioxidants (Basel) 2023; 12:1331. [PMID: 37507871 PMCID: PMC10376326 DOI: 10.3390/antiox12071331] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Oxidative stress surrounding cancer cells provides them with certain growth and survival advantages necessary for disease progression. In this context, Se-containing molecules have gained attention due to their anticancer and antioxidant activity. In our previous work, we synthesized a library of 39 selenoesters containing functional groups commonly present in natural products (NP), which showed potent anticancer activity, but did not demonstrate high radical scavenger activity. Thus, 20 novel Se derivatives resembling NP have been synthesized presenting acylselenourea functionality in their structures. Radical scavenger activity was tested using DPPH assay and in vitro protective effects against ROS-induced cell death caused by H2O2. Additionally, antiproliferative activity was evaluated in prostate, colon, lung, and breast cancer cell lines, along with their ability to induce apoptosis. Compounds 1.I and 5.I showed potent cytotoxicity against the tested cancer cell lines, along with high selectivity indexes and induction of caspase-mediated apoptosis. These compounds exhibited potent and concentration-dependent radical scavenging activity achieving DPPH inhibition similar to ascorbic acid and trolox. To conclude, we have demonstrated that the introduction of Se in the form of acylselenourea into small molecules provides strong radical scavengers in vitro and antiproliferative activity, which may lead to the development of promising dual compounds.
Collapse
Affiliation(s)
- Nora Astrain-Redin
- Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea 1, 31008 Pamplona, Spain
- Department of Pharmacology, Penn State Cancer Institute, CH72, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Asif Raza
- Department of Pharmacology, Penn State Cancer Institute, CH72, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Ignacio Encío
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, 31008 Pamplona, Spain
- Departamento de Ciencias de la Salud, Universidad Pública de Navarra, Avda. Barañain s/n, 31008 Pamplona, Spain
| | - Arun K Sharma
- Department of Pharmacology, Penn State Cancer Institute, CH72, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Daniel Plano
- Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea 1, 31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, 31008 Pamplona, Spain
| | - Carmen Sanmartín
- Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea 1, 31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, 31008 Pamplona, Spain
| |
Collapse
|
7
|
Amin A, Lone A, Farooq F, Wani UM, Kawoosa F, Qadri RA. Identification of novel inhibitors of tetranectin-plasminogen interaction to suppress breast cancer invasion: an integrated computational and cell-based investigation. J Biomol Struct Dyn 2023; 41:15023-15032. [PMID: 36927470 DOI: 10.1080/07391102.2023.2187228] [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: 11/03/2022] [Accepted: 02/22/2023] [Indexed: 03/18/2023]
Abstract
Tetranectin-plasminogen interaction plays a defining role in extracellular matrix degradation, enabling tumor cell invasion and metastasis. This interaction occurs via the carbohydrate recognition domain (CRD) and Kringle 4 domain of tetranectin and plasminogen, respectively, leading to activation of the plasminogen-cascade that triggers the proteolytic processes. Thus targeting this interaction represents an important strategy to suppress tumor cell migration and invasion. In this direction, we attempted to target the CRD of tetranectin to inhibit its interaction with the Kringle-4 domain of plasminogen using natural bioactive compounds. A cheminformatics pipeline for drug designing and screening was utilized to obtain lead compound(s) that exhibit conformationally and energetically viable CRD binding. Out of 206 compounds screened, diosgenin and scytonemin displayed the most favorable interactions with CRD. Short-term molecular dynamics simulations of 20 ns were employed to further study the conformational stability of both compounds with tetranectin CRD which reflected at the increased stability of diosgenin in the CRD binding pocket compared to scytonemin. Finally, an extended molecular dynamic simulation of 100 ns affirmed the robust and stable interaction of diosgenin with CRD. Furthermore, diosgenin was observed to exert a pronounced anti-proliferative effect on high tetranectin-expressing MDA-MB-231 breast cancer cells. The inhibitory effect of diosgenin on the tetranectin-plasminogen interaction was corroborated by the reduced migration and invasiveness of MDA-MB-231 cells under diosgenin treatment. Overall the study presents an alternate and safer approach to impede breast cancer metastasis and delineates the novel anti-metastatic activity of diosgenin.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Asif Amin
- Immunobiology Lab, Department of Biotechnology, University of Kashmir, Srinagar, J&K, India
| | - Asif Lone
- Department of Biochemistry, Deshbandhu College, University of Delhi, Delhi, India
| | - Faizah Farooq
- Immunobiology Lab, Department of Biotechnology, University of Kashmir, Srinagar, J&K, India
| | - Umer Majeed Wani
- Immunobiology Lab, Department of Biotechnology, University of Kashmir, Srinagar, J&K, India
| | - Fizallah Kawoosa
- Department of Immunology and Molecular Medicine, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, J&K, India
| | - Raies A Qadri
- Immunobiology Lab, Department of Biotechnology, University of Kashmir, Srinagar, J&K, India
| |
Collapse
|
8
|
Huang Y, Peng Z, Wei M, Pang L, Cheng Y, Xiao JA, Gan C, Cui J. Straightforward synthesis of steroidal selenocyanates through oxidative umpolung selenocyanation of steroids and their antitumor activity. J Steroid Biochem Mol Biol 2023; 225:106203. [PMID: 36228841 DOI: 10.1016/j.jsbmb.2022.106203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 11/07/2022]
Abstract
Straightforward access to steroidal selenocyanates in a single assembly step from steroids remains a significant challenge. However, the development of novel method for the synthesis of steroidal selenocyanates and further investigation of their bioactivities have largely lagged behind. In this work, selenocyano groups were directly introduced into the 17- or 21-position of pregnenolone, the 2-position of estradiol, and the 16-position of estrone. A total of 16 estrogen selenocyanate derivatives with diverse structures were synthesized, and the tumor cell lines closely related to the expression level of estrogen were used to investigate the inhibitory activity of the target products on tumor cell proliferation in vitro. The results revealed that the 17-selenocyano-substituted pregnenolone selenocyanate derivatives 1b-3b exhibit obvious inhibitory activity against the tested tumor cell lines. Additionally, the 2-selenocyano-substituted estradiol derivatives and 16-selenocyano-substituted estrone derivatives exhibit selective inhibitory on HeLa cell lines. Among them, 2-selenocyano-3-methoxyestradiol-17-benzoate (7e) displayed an IC50 value of 4.1 µM against HeLa cells and induced programmed apoptosis in HeLa cancer cells. Furthermore, compound 7e could significantly inhibit the growth of human cervical cancer xenografts in zebrafish in vivo. This approach provides new insights for future steroid antitumor drug design.
Collapse
Affiliation(s)
- Yanmin Huang
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China
| | - Zining Peng
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China
| | - Meizhen Wei
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China
| | - Liping Pang
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China
| | - Yang Cheng
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China
| | - Jun-An Xiao
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China.
| | - Chunfang Gan
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China
| | - Jianguo Cui
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China.
| |
Collapse
|
9
|
Bansal R, Suryan A. A Comprehensive Review on Steroidal Bioconjugates as Promising Leads in Drug Discovery. ACS BIO & MED CHEM AU 2022; 2:340-369. [PMID: 37102169 PMCID: PMC10125316 DOI: 10.1021/acsbiomedchemau.1c00071] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Ever increasing unmet medical requirements of the human race and the continuous fight for survival against variety of diseases give birth to novel molecules through research. As diseases evolve, different strategies are employed to counter the new challenges and to discover safer, more effective, and target-specific therapeutic agents. Among several novel approaches, bioconjugation, in which two chemical moieties are joined together to achieve noticeable results, has emerged as a simple and convenient technique for a medicinal chemist to obtain potent molecules. The steroid system has been extensively used as a privileged scaffold gifted with significantly diversified medicinal properties in the drug discovery and development process. Steroidal molecules are preferred for their rigidness and good ability to penetrate biological membranes. Slight alteration in the basic ring structure results in the formation of steroidal derivatives with a wide range of therapeutic activities. Steroids are not only active as such, conjugating them with various biologically active moieties results in increased lipophilicity, stability, and target specificity with decreased adverse effects. Thus, the steroid nucleus prominently behaves as a biological carrier for small molecules. The steroid bioconjugates offer several advantages such as synergistic activity with fewer side effects due to reduced dose and selective therapy. The steroidal bioconjugates have been widely explored for their usefulness against various disorders and have shown significant utility as anticancer, anti-inflammatory, anticoagulant, antimicrobial, insecticidal/pesticidal, antioxidant, and antiviral agents along with several other miscellaneous activities. This work provides a comprehensive review on the therapeutic progression of steroidal bioconjugates as medicinally active molecules. The review covers potential biological applications of steroidal bioconjugates and would benefit the wider scientific community in their drug discovery endeavors.
Collapse
Affiliation(s)
- Ranju Bansal
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India
| | - Amruta Suryan
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India
| |
Collapse
|
10
|
Wang D, Wang X. Diosgenin and Its Analogs: Potential Protective Agents Against Atherosclerosis. Drug Des Devel Ther 2022; 16:2305-2323. [PMID: 35875677 PMCID: PMC9304635 DOI: 10.2147/dddt.s368836] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/09/2022] [Indexed: 11/23/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the artery wall associated with lipid metabolism imbalance and maladaptive immune response, which mediates most cardiovascular events. First-line drugs such as statins and antiplatelet drug aspirin have shown good effects against atherosclerosis but may lead to certain side effects. Thus, the development of new, safer, and less toxic agents for atherosclerosis is urgently needed. Diosgenin and its analogs have gained importance for their efficacy against life-threatening diseases, including cardiovascular, endocrine, nervous system diseases, and cancer. Diosgenin and its analogs are widely found in the rhizomes of Dioscore, Solanum, and other species and share similar chemical structures and pharmacological effects. Recent data suggested diosgenin plays an anti-atherosclerosis role through its anti-inflammatory, antioxidant, plasma cholesterol-lowering, anti-proliferation, and anti-thrombotic effects. However, a review of the effects of diosgenin and its natural structure analogs on AS is still lacking. This review summarizes the effects of diosgenin and its analogs on vascular endothelial dysfunction, vascular smooth muscle cell (VSMC) proliferation, migration and calcification, lipid metabolism, and inflammation, and provides a new overview of its anti-atherosclerosis mechanism. Besides, the structures, sources, safety, pharmacokinetic characteristics, and biological availability are introduced to reveal the limitations and challenges of current studies, hoping to provide a theoretical basis for the clinical application of diosgenin and its analogs and provide a new idea for developing new agents for atherosclerosis.
Collapse
Affiliation(s)
- Dan Wang
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai, People’s Republic of China
| | - Xiaolong Wang
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai, People’s Republic of China
- Correspondence: Xiaolong Wang, Tel +86 13501991450, Fax +86 21 51322445, Email
| |
Collapse
|
11
|
Erdagi SI, Yildiz U. Synthesis, Structural Analysis and Antiproliferative Activity of Nitrogen‐Containing Hetero Spirostan Derivatives: Oximes, Heterocyclic Ring‐Fused and Furostanes. ChemistrySelect 2022. [DOI: 10.1002/slct.202200439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Sevinc Ilkar Erdagi
- Department of Chemistry Kocaeli University Umuttepe campus 41380 Kocaeli Turkey
| | - Ufuk Yildiz
- Department of Chemistry Kocaeli University Umuttepe campus 41380 Kocaeli Turkey
| |
Collapse
|
12
|
Lissette Mora-Medina T, Martínez-Pascual R, Ángel Peña-Rico M, Viñas-Bravo O, Montiel-Smith S, Pérez-Picaso L, Moreno-Díaz H. Preparation and cytotoxic evaluation of new steroidal oximes and aza-homosteroids from diosgenin and cholesterol. Steroids 2022; 182:109012. [PMID: 35307325 DOI: 10.1016/j.steroids.2022.109012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/08/2022] [Accepted: 03/14/2022] [Indexed: 11/27/2022]
Abstract
Using cholesterol and diosgenin as starting materials, we have designed a straightforward methodology to prepare in a reduced number of steps a novel series of steroidal oximes and their aza-homolactam analogs with four types of side chains: cholestane, spirostane, 22-oxocholestane and 22,26-epoxycholestene. The products were evaluated for their cytotoxic activity against the MCF-7 breast cancer cell line. Moreover, the selectivity of the most active compounds was determined against peripheral blood lymphocytes. Compounds 5, 8 and 13 were found to be the most active derivatives, exhibiting IC50 values in the low micromolar range (7.9-9.5 µM) and excellent selectivities (IC50 > 100 µM) against the non-tumor cell line.
Collapse
Affiliation(s)
- Thalía Lissette Mora-Medina
- División de Estudios de Posgrado, Maestría en Ciencias Químicas, Universidad del Papaloapan, Circuito Central 200, Col. Parque Industrial, Tuxtepec, 68301 Oaxaca, Mexico
| | - Roxana Martínez-Pascual
- Centro de Investigaciones Científicas, Instituto de Química Aplicada, Universidad del Papaloapan, Circuito Central 200, Col. Parque Industrial, Tuxtepec, 68301 Oaxaca, Mexico.
| | - Miguel Ángel Peña-Rico
- Centro de Investigaciones Científicas, Instituto de Química Aplicada, Universidad del Papaloapan, Circuito Central 200, Col. Parque Industrial, Tuxtepec, 68301 Oaxaca, Mexico
| | - Omar Viñas-Bravo
- Centro de Investigaciones Científicas, Instituto de Química Aplicada, Universidad del Papaloapan, Circuito Central 200, Col. Parque Industrial, Tuxtepec, 68301 Oaxaca, Mexico
| | - Sara Montiel-Smith
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, C.P. 72570, Puebla, Pue., Mexico
| | - Lemuel Pérez-Picaso
- Centro de Investigaciones Científicas, Instituto de Química Aplicada, Universidad del Papaloapan, Circuito Central 200, Col. Parque Industrial, Tuxtepec, 68301 Oaxaca, Mexico
| | - Hermenegilda Moreno-Díaz
- Centro de Investigaciones Científicas, Instituto de Química Aplicada, Universidad del Papaloapan, Circuito Central 200, Col. Parque Industrial, Tuxtepec, 68301 Oaxaca, Mexico
| |
Collapse
|
13
|
Synthesis of diosgenin derivatives by A and B ring modifications and low-valent titanium (Ti0)-catalysed McMurry coupling reactions and designing to create novel biological agents. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
14
|
Fan R, He W, Fan Y, Xu W, Xu W, Yan G, Xu S. Recent advances in chemical synthesis, biocatalysis, and biological evaluation of diosgenin derivatives - A review. Steroids 2022; 180:108991. [PMID: 35217033 DOI: 10.1016/j.steroids.2022.108991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 01/19/2022] [Accepted: 02/18/2022] [Indexed: 12/28/2022]
Abstract
Extracting organic compounds from plants and developing derivatives are essential methods for drug discovery. Diosgenin, extracted from Dioscoreaceae plants, is a type of spirostan steroid with various biological effects, including anti-inflammation, neuro-protection, and apoptosis-induction. Many researchers committed their work to the chemical semi-synthesis of diosgenin derivatives to improve diosgenin's therapeutic bioavailability and expand its range of applications in disease treatment and prevention. Biotransformation, a mild whole-cell biocatalysis method, also made crucial contributions to the structural diversity of diosgenin analogs in recent years. Although the structural modification of diosgenin has made significant progress, it lacks a comprehensive review. Here, we review the chemical modification and biotransformation of diosgenin along with the biological evaluation of diosgenin derivatives to provide a reference for the structural modification strategy and pharmaceutical application of diosgenin derivatives.
Collapse
Affiliation(s)
- Ruolan Fan
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | - Weishen He
- Biology Department, Boston College, Brighton, MA 02135, USA
| | - Yong Fan
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | - Wen Xu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | - Wei Xu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China.
| | - Guohong Yan
- Pharmacy Department, People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350004, PR China.
| | - Shaohua Xu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China.
| |
Collapse
|
15
|
Fernández Y, Movellan J, Foradada L, Giménez V, García‐Aranda N, Mancilla S, Armiñán A, Borgos SE, Hyldbakk A, Bogdanska A, Gobbo OL, Prina‐Mello A, Ponti J, Calzolai L, Zagorodko O, Gallon E, Niño‐Pariente A, Paul A, Schwartz Jr S, Abasolo I, Vicent MJ. In Vivo Antitumor and Antimetastatic Efficacy of a Polyacetal-Based Paclitaxel Conjugate for Prostate Cancer Therapy. Adv Healthc Mater 2022; 11:e2101544. [PMID: 34706167 DOI: 10.1002/adhm.202101544] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/25/2021] [Indexed: 12/12/2022]
Abstract
Prostate cancer (PCa), one of the leading causes of cancer-related deaths, currently lacks effective treatment for advanced-stage disease. Paclitaxel (PTX) is a highly active chemotherapeutic drug and the first-line treatment for PCa; however, conventional PTX formulation causes severe hypersensitivity reactions and limits PTX use at high concentrations. In the pursuit of high molecular weight, biodegradable, and pH-responsive polymeric carriers, one conjugates PTX to a polyacetal-based nanocarrier to yield a tert-Ser-PTX polyacetal conjugate. tert-Ser-PTX conjugate provides sustained release of PTX over 2 weeks in a pH-responsive manner while also obtaining a degree of epimerization of PTX to 7-epi-PTX. Serum proteins stabilize tert-Ser-PTX, with enhanced stability in human serum versus PBS (pH 7.4). In vitro efficacy assessments in PCa cells demonstrate IC50 values above those for the free form of PTX due to the differential cell trafficking modes; however, in vivo tolerability assays demonstrate that tert-Ser-PTX significantly reduces the systemic toxicities associated with free PTX treatment. tert-Ser-PTX also effectively inhibits primary tumor growth and hematologic, lymphatic, and coelomic dissemination, as confirmed by in vivo and ex vivo bioluminescence imaging and histopathological evaluations in mice carrying orthotopic LNCaP tumors. Overall, the results suggest the application of tert-Ser-PTX as a robust antitumor/antimetastatic treatment for PCa.
Collapse
Affiliation(s)
- Yolanda Fernández
- Functional Validation & Preclinical Research (FVPR) CIBBIM‐Nanomedicine Vall d'Hebron Institut de Recerca (VHIR) Universitat Autònoma de Barcelona (UAB) Barcelona 08035 Spain
- Drug Delivery & Targeting Group CIBBIM‐Nanomedicine Vall d'Hebron Institut de Recerca (VHIR) Universitat Autònoma de Barcelona (UAB) Barcelona 08035 Spain
- Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER‐BBN) Barcelona 08035 Spain
| | - Julie Movellan
- Polymer Therapeutics Laboratory. Centro de Investigación Príncipe Felipe. Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| | - Laia Foradada
- Functional Validation & Preclinical Research (FVPR) CIBBIM‐Nanomedicine Vall d'Hebron Institut de Recerca (VHIR) Universitat Autònoma de Barcelona (UAB) Barcelona 08035 Spain
- Drug Delivery & Targeting Group CIBBIM‐Nanomedicine Vall d'Hebron Institut de Recerca (VHIR) Universitat Autònoma de Barcelona (UAB) Barcelona 08035 Spain
- Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER‐BBN) Barcelona 08035 Spain
| | - Vanessa Giménez
- Polymer Therapeutics Laboratory. Centro de Investigación Príncipe Felipe. Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| | - Natalia García‐Aranda
- Functional Validation & Preclinical Research (FVPR) CIBBIM‐Nanomedicine Vall d'Hebron Institut de Recerca (VHIR) Universitat Autònoma de Barcelona (UAB) Barcelona 08035 Spain
- Drug Delivery & Targeting Group CIBBIM‐Nanomedicine Vall d'Hebron Institut de Recerca (VHIR) Universitat Autònoma de Barcelona (UAB) Barcelona 08035 Spain
- Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER‐BBN) Barcelona 08035 Spain
| | - Sandra Mancilla
- Functional Validation & Preclinical Research (FVPR) CIBBIM‐Nanomedicine Vall d'Hebron Institut de Recerca (VHIR) Universitat Autònoma de Barcelona (UAB) Barcelona 08035 Spain
- Drug Delivery & Targeting Group CIBBIM‐Nanomedicine Vall d'Hebron Institut de Recerca (VHIR) Universitat Autònoma de Barcelona (UAB) Barcelona 08035 Spain
- Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER‐BBN) Barcelona 08035 Spain
| | - Ana Armiñán
- Polymer Therapeutics Laboratory. Centro de Investigación Príncipe Felipe. Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| | - Sven Even Borgos
- Department of Biotechnology and Nanomedicine SINTEF Industry Trondheim NO‐7465 Norway
| | - Astrid Hyldbakk
- Department of Biotechnology and Nanomedicine SINTEF Industry Trondheim NO‐7465 Norway
| | - Anna Bogdanska
- Laboratory for Biological Characterization of Advanced Materials (LBCAM) Trinity Translational Medicine Institute Trinity College Dublin Dublin D08 W9RT Ireland
- Trinity St James's Cancer Institute Trinity College Dublin the University of Dublin Dublin D08 W9RT Ireland
| | - Oliviero L. Gobbo
- Trinity St James's Cancer Institute Trinity College Dublin the University of Dublin Dublin D08 W9RT Ireland
- School of Pharmacy and Pharmaceutical Sciences Trinity College Dublin Dublin D02 R590 Ireland
| | - Adriele Prina‐Mello
- Laboratory for Biological Characterization of Advanced Materials (LBCAM) Trinity Translational Medicine Institute Trinity College Dublin Dublin D08 W9RT Ireland
- Trinity St James's Cancer Institute Trinity College Dublin the University of Dublin Dublin D08 W9RT Ireland
| | - Jessica Ponti
- European Commission Joint Research Centre (JRC) via Fermi 2749 Ispra 21027 Italy
| | - Luigi Calzolai
- European Commission Joint Research Centre (JRC) via Fermi 2749 Ispra 21027 Italy
| | - Oleksandr Zagorodko
- Polymer Therapeutics Laboratory. Centro de Investigación Príncipe Felipe. Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| | - Elena Gallon
- Polymer Therapeutics Laboratory. Centro de Investigación Príncipe Felipe. Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| | - Amaya Niño‐Pariente
- Polymer Therapeutics Laboratory. Centro de Investigación Príncipe Felipe. Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| | - Alison Paul
- School of Chemistry Cardiff University Main Building, Park Place Cardiff CF10 3AT UK
| | - Simó Schwartz Jr
- Drug Delivery & Targeting Group CIBBIM‐Nanomedicine Vall d'Hebron Institut de Recerca (VHIR) Universitat Autònoma de Barcelona (UAB) Barcelona 08035 Spain
- Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER‐BBN) Barcelona 08035 Spain
| | - Ibane Abasolo
- Functional Validation & Preclinical Research (FVPR) CIBBIM‐Nanomedicine Vall d'Hebron Institut de Recerca (VHIR) Universitat Autònoma de Barcelona (UAB) Barcelona 08035 Spain
- Drug Delivery & Targeting Group CIBBIM‐Nanomedicine Vall d'Hebron Institut de Recerca (VHIR) Universitat Autònoma de Barcelona (UAB) Barcelona 08035 Spain
- Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER‐BBN) Barcelona 08035 Spain
| | - María J. Vicent
- Polymer Therapeutics Laboratory. Centro de Investigación Príncipe Felipe. Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| |
Collapse
|
16
|
Simple Zn-Mediated Seleno- and Thio-Functionalization of Steroids at C-1 Position. Int J Mol Sci 2022; 23:ijms23063022. [PMID: 35328446 PMCID: PMC8952209 DOI: 10.3390/ijms23063022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 11/17/2022] Open
Abstract
Here we report the reaction in the biphasic system of the in situ prepared selenols and thiols with 1,4-androstadiene-3,17-dione (1) or prednisone acetate (2) having α,β-unsaturated ketone as an electrophilic functionalization. The Michael-type addition reaction resulted to be chemo- and stereoselective, affording a series of novel steroidal selenides and sulfides. This is an example of a one-step, eco-friendly process that bypasses some of the main concerns connected with the bad smell and the toxicity of these seleno- and thio-reagents. Furthermore, we demonstrated that the proposed methodology offers the possibility to prepare libraries of steroids variously and selectively decorated with different organochalcogen moieties at the C1 position starting from 1,4-androstadienic skeletons and leaving unaltered the C4-C5 unsaturation. Based on the data reported in the literature the introduction of an organoselenium or an organosulfur moiety in a steroid could provide new interesting pharmaceutically active entities exerting anticancer and antimicrobial activities. In this optic, new synthetic strategies to efficiently prepare this class of compounds could be strongly desirable.
Collapse
|
17
|
Hou W, Xu H. Incorporating Selenium into Heterocycles and Natural Products─From Chemical Properties to Pharmacological Activities. J Med Chem 2022; 65:4436-4456. [PMID: 35244394 DOI: 10.1021/acs.jmedchem.1c01859] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Selenium (Se)-containing compounds have emerged as potential therapeutic agents for the treatment of a range of diseases. Through tremendous effort, considerable knowledge has been acquired to understand the complex chemical properties and biological activities of selenium, especially after its incorporation into bioactive molecules. From this perspective, we compiled extensive literature evidence to summarize and critically discuss the relationship between the pharmacological activities and chemical properties of selenium compounds and the strategic incorporation of selenium into organic molecules, especially bioactive heterocycles and natural products. We also provide perspectives regarding the challenges in selenium-based medicinal chemistry and future research directions.
Collapse
Affiliation(s)
- Wei Hou
- College of Pharmaceutical Science and Institute of Drug Development and Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hongtao Xu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
18
|
Ibáñez-Escribano A, Fonseca-Berzal C, Martínez-Montiel M, Álvarez-Márquez M, Gómez-Núñez M, Lacueva-Arnedo M, Espinosa-Buitrago T, Martín-Pérez T, Escario JA, Merino-Montiel P, Montiel-Smith S, Gómez-Barrio A, López Ó, Fernández-Bolaños JG. Thio- and selenosemicarbazones as antiprotozoal agents against Trypanosoma cruzi and Trichomonas vaginalis. J Enzyme Inhib Med Chem 2022; 37:781-791. [PMID: 35193444 PMCID: PMC8881069 DOI: 10.1080/14756366.2022.2041629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Herein, we report the preparation of a panel of Schiff bases analogues as antiprotozoal agents by modification of the stereoelectronic effects of the substituents on N-1 and N-4 and the nature of the chalcogen atom (S, Se). These compounds were evaluated towards Trypanosoma cruzi and Trichomonas vaginalis. Thiosemicarbazide 31 showed the best trypanocidal profile (epimastigotes), similar to benznidazole (BZ): IC50 (31)=28.72 μM (CL-B5 strain) and 33.65 μM (Y strain), IC50 (BZ)=25.31 μM (CL-B5) and 22.73 μM (Y); it lacked toxicity over mammalian cells (CC50 > 256 µM). Thiosemicarbazones 49, 51 and 63 showed remarkable trichomonacidal effects (IC50 =16.39, 14.84 and 14.89 µM) and no unspecific cytotoxicity towards Vero cells (CC50 ≥ 275 µM). Selenoisosters 74 and 75 presented a slightly enhanced activity (IC50=11.10 and 11.02 µM, respectively). Hydrogenosome membrane potential and structural changes were analysed to get more insight into the trichomonacidal mechanism.
Collapse
Affiliation(s)
- Alexandra Ibáñez-Escribano
- Unidad de Parasitología, Departamento de Microbiología y Parasitología, Facultad de Farmacia, Madrid, Spain
| | - Cristina Fonseca-Berzal
- Unidad de Parasitología, Departamento de Microbiología y Parasitología, Facultad de Farmacia, Madrid, Spain
| | - Mónica Martínez-Montiel
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Manuel Álvarez-Márquez
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Sevilla, Spain
| | - María Gómez-Núñez
- Escuela Politécnica Superior, Universidad de Sevilla, Sevilla, Spain
| | - Manuel Lacueva-Arnedo
- Unidad de Parasitología, Departamento de Microbiología y Parasitología, Facultad de Farmacia, Madrid, Spain
| | - Teresa Espinosa-Buitrago
- Unidad de Parasitología, Departamento de Microbiología y Parasitología, Facultad de Farmacia, Madrid, Spain
| | - Tania Martín-Pérez
- Departamento de Biomedicina y Biotecnología, Facultad de Farmacia, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain.,Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - José Antonio Escario
- Unidad de Parasitología, Departamento de Microbiología y Parasitología, Facultad de Farmacia, Madrid, Spain
| | - Penélope Merino-Montiel
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Sara Montiel-Smith
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Alicia Gómez-Barrio
- Unidad de Parasitología, Departamento de Microbiología y Parasitología, Facultad de Farmacia, Madrid, Spain
| | - Óscar López
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Sevilla, Spain
| | | |
Collapse
|
19
|
Li Y, Wang K, Chen Y, Cai J, Qin X, Lu A, Guan D, Qin G, Chen W. A System Pharmacology Model for Decoding the Synergistic Mechanisms of Compound Kushen Injection in Treating Breast Cancer. Front Pharmacol 2021; 12:723147. [PMID: 34899291 PMCID: PMC8660088 DOI: 10.3389/fphar.2021.723147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/15/2021] [Indexed: 11/29/2022] Open
Abstract
Breast cancer (BC) is one of the most common malignant tumors among women worldwide and can be treated using various methods; however, side effects of these treatments cannot be ignored. Increasing evidence indicates that compound kushen injection (CKI) can be used to treat BC. However, traditional Chinese medicine (TCM) is characterized by “multi-components” and “multi-targets”, which make it challenging to clarify the potential therapeutic mechanisms of CKI on BC. Herein, we designed a novel system pharmacology strategy using differentially expressed gene analysis, pharmacokinetics synthesis screening, target identification, network analysis, and docking validation to construct the synergy contribution degree (SCD) and therapeutic response index (TRI) model to capture the critical components responding to synergistic mechanisms of CKI in BC. Through our designed mathematical models, we defined 24 components as a high contribution group of synergistic components (HCGSC) from 113 potentially active components of CKI based on ADME parameters. Pathway enrichment analysis of HCGSC targets indicated that Rhizoma Heterosmilacis and Radix Sophorae Flavescentis could synergistically target the PI3K-Akt signaling pathway and the cAMP signaling pathway to treat BC. Additionally, TRI analysis showed that the average affinity of HCGSC and targets involved in the key pathways reached -6.47 kcal/mmol, while in vitro experiments proved that two of the three high TRI-scored components in the HCGSC showed significant inhibitory effects on breast cancer cell proliferation and migration. These results demonstrate the accuracy and reliability of the proposed strategy.
Collapse
Affiliation(s)
- Yi Li
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kexin Wang
- Institute of Integrated Bioinformedicine and Translational Science, Hong Kong Baptist University, Hong Kong SAR, China.,Neurosurgery Center, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yupeng Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Key Laboratory of Biochip Technology, Southern Medical University, Guangzhou, China
| | - Jieqi Cai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Key Laboratory of Biochip Technology, Southern Medical University, Guangzhou, China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China
| | - Aiping Lu
- Institute of Integrated Bioinformedicine and Translational Science, Hong Kong Baptist University, Hong Kong SAR, China
| | - Daogang Guan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Key Laboratory of Biochip Technology, Southern Medical University, Guangzhou, China
| | - Genggeng Qin
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weiguo Chen
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Liao J, Liao G, Gao Y, Chai X, Wu Q, Zhao Q. Synthesis and Biological Activities of Diosgenin-Triazole Conjugates with a 1,3-Dipolar Cycloaddition Reaction. Chem Nat Compd 2021. [DOI: 10.1007/s10600-021-03552-2] [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]
|
21
|
Jastrzebska I, Grzes PA, Niemirowicz-Laskowska K, Car H. Selenosteroids - promising hybrid compounds with pleiotropic biological activity: synthesis and biological aspects. J Steroid Biochem Mol Biol 2021; 213:105975. [PMID: 34418527 DOI: 10.1016/j.jsbmb.2021.105975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 01/22/2023]
Abstract
It is established that steroid based agents are an example of compounds obtained from natural patterns and are of great importance due to their application in the prevention and treatment of diseases. Selenosteroids are hybrids formed by attaching Se-moiety to a steroid molecule. In these types of hybrids, selenium can be present as selenide or as a part of selenosemicarbazones, isoselenocyanates, selenourea, etc. Attaching a Se-moiety to a biologically active steroid might enhance the biological properties of both fragments. Available literature indicates that these kinds of hybrids demonstrate significant anticancer activity, which renders them interesting in terms of medical use. In this review, we present various methods of synthesis and demonstrate that seleno-steroid compounds are promising molecules for further pharmaceutical application.
Collapse
Affiliation(s)
- Izabella Jastrzebska
- Faculty of Chemistry, University of Białystok, ul. Ciołkowskiego 1K, 15-245, Białystok, Poland.
| | - Pawel A Grzes
- Faculty of Chemistry, University of Białystok, ul. Ciołkowskiego 1K, 15-245, Białystok, Poland
| | | | - Halina Car
- Department of Experimental Pharmacology, Medical University of Bialystok, ul. Szpitalna 37, 15-295, Białystok, Poland
| |
Collapse
|
22
|
Harvey DJ. ANALYSIS OF CARBOHYDRATES AND GLYCOCONJUGATES BY MATRIX-ASSISTED LASER DESORPTION/IONIZATION MASS SPECTROMETRY: AN UPDATE FOR 2015-2016. MASS SPECTROMETRY REVIEWS 2021; 40:408-565. [PMID: 33725404 DOI: 10.1002/mas.21651] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/24/2020] [Indexed: 06/12/2023]
Abstract
This review is the ninth update of the original article published in 1999 on the application of matrix-assisted laser desorption/ionization (MALDI) mass spectrometry to the analysis of carbohydrates and glycoconjugates and brings coverage of the literature to the end of 2016. Also included are papers that describe methods appropriate to analysis by MALDI, such as sample preparation techniques, even though the ionization method is not MALDI. Topics covered in the first part of the review include general aspects such as theory of the MALDI process, matrices, derivatization, MALDI imaging, fragmentation and arrays. The second part of the review is devoted to applications to various structural types such as oligo- and poly-saccharides, glycoproteins, glycolipids, glycosides and biopharmaceuticals. Much of this material is presented in tabular form. The third part of the review covers medical and industrial applications of the technique, studies of enzyme reactions and applications to chemical synthesis. The reported work shows increasing use of combined new techniques such as ion mobility and the enormous impact that MALDI imaging is having. MALDI, although invented over 30 years ago is still an ideal technique for carbohydrate analysis and advancements in the technique and range of applications show no sign of deminishing. © 2020 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David J Harvey
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, United Kingdom
| |
Collapse
|
23
|
Agrahari AK, Bose P, Jaiswal MK, Rajkhowa S, Singh AS, Hotha S, Mishra N, Tiwari VK. Cu(I)-Catalyzed Click Chemistry in Glycoscience and Their Diverse Applications. Chem Rev 2021; 121:7638-7956. [PMID: 34165284 DOI: 10.1021/acs.chemrev.0c00920] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Copper(I)-catalyzed 1,3-dipolar cycloaddition between organic azides and terminal alkynes, commonly known as CuAAC or click chemistry, has been identified as one of the most successful, versatile, reliable, and modular strategies for the rapid and regioselective construction of 1,4-disubstituted 1,2,3-triazoles as diversely functionalized molecules. Carbohydrates, an integral part of living cells, have several fascinating features, including their structural diversity, biocompatibility, bioavailability, hydrophilicity, and superior ADME properties with minimal toxicity, which support increased demand to explore them as versatile scaffolds for easy access to diverse glycohybrids and well-defined glycoconjugates for complete chemical, biochemical, and pharmacological investigations. This review highlights the successful development of CuAAC or click chemistry in emerging areas of glycoscience, including the synthesis of triazole appended carbohydrate-containing molecular architectures (mainly glycohybrids, glycoconjugates, glycopolymers, glycopeptides, glycoproteins, glycolipids, glycoclusters, and glycodendrimers through regioselective triazole forming modular and bio-orthogonal coupling protocols). It discusses the widespread applications of these glycoproducts as enzyme inhibitors in drug discovery and development, sensing, gelation, chelation, glycosylation, and catalysis. This review also covers the impact of click chemistry and provides future perspectives on its role in various emerging disciplines of science and technology.
Collapse
Affiliation(s)
- Anand K Agrahari
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Priyanka Bose
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Manoj K Jaiswal
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Sanchayita Rajkhowa
- Department of Chemistry, Jorhat Institute of Science and Technology (JIST), Jorhat, Assam 785010, India
| | - Anoop S Singh
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Srinivas Hotha
- Department of Chemistry, Indian Institute of Science and Engineering Research (IISER), Pune, Maharashtra 411021, India
| | - Nidhi Mishra
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Vinod K Tiwari
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
24
|
Chuai H, Zhang SQ, Bai H, Li J, Wang Y, Sun J, Wen E, Zhang J, Xin M. Small molecule selenium-containing compounds: Recent development and therapeutic applications. Eur J Med Chem 2021; 223:113621. [PMID: 34217061 DOI: 10.1016/j.ejmech.2021.113621] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023]
Abstract
Selenium (Se) is an essential micronutrient of organism and has important function. It participates in the functions of selenoprotein in several manners. In recent years, Se has attracted much attention because of its therapeutic potential against several diseases. Many natural and synthetic organic Se-containing compounds were studied and explored for the treatment of cancer and other diseases. Studies have showed that incorporation of Se atom into small molecules significantly enhanced their bioactivities. In this paper, according to different applications and structural characteristics, the research progress and therapeutic application of Se-containing compounds are reviewed, and more than 110 Se-containing compounds were selected as representatives which showed potent activities such as anticancer, antioxidant, antifibrolytic, antiparasitic, antibacterial, antiviral, antifungal and central nervous system related effects. This review is expected to provide a basis for further study of new promising Se-containing compounds.
Collapse
Affiliation(s)
- Hongyan Chuai
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, Shaanxi, 710061, PR China
| | - San-Qi Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, Shaanxi, 710061, PR China
| | - Huanrong Bai
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, Shaanxi, 710061, PR China
| | - Jiyu Li
- Henan Xibaikang Health Industry Co., Ltd, Jiyuan, Henan, 459006, PR China
| | - Yang Wang
- Henan Xibaikang Health Industry Co., Ltd, Jiyuan, Henan, 459006, PR China
| | - Jiajia Sun
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, Shaanxi, 710061, PR China
| | - Ergang Wen
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, Shaanxi, 710061, PR China
| | - Jiye Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, Shaanxi, 710061, PR China
| | - Minhang Xin
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, Shaanxi, 710061, PR China.
| |
Collapse
|
25
|
Shahrajabian MH, Sun W, Marmitt DJ, Cheng Q. Diosgenin and galactomannans, natural products in the pharmaceutical sciences. CLINICAL PHYTOSCIENCE 2021. [DOI: 10.1186/s40816-021-00288-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Diosgenin is an isospirostane derivative, which is a steroidal sapogenin and the product of acids or enzymes hydrolysis process of dioscin and protodioscin. Galactomannans are heteropolysaccharides composed of D-mannose and D-galactose, which are major sources of locust bean, guar, tara and fenugreek.
Methods
Literature survey was accomplished using multiple databases including PubMed, Science Direct, ISI web of knowledge and Google Scholar.
Results
Four major sources of seed galactomannans are locust bean (Ceratonia siliqua), guar (Cyamopsis tetragonoloba), tara (Caesalpinia spinosa Kuntze), and fenugreek (T.foenum-graecum). Diosgenin has effect on immune system, lipid system, inflammatory and reproductive systems, caner, metabolic process, blood system, blood glucose and calcium regulation. The most important pharmacological benefits of galactomannan are antidiabetic, antioxidant, anticancer, anticholinesterase, antiviral activities, and appropriate for dengue virus and gastric diseases.
Conclusions
Considering the importance of diosgenin and galactomannans, the obtained findings suggest potential of diosgenin and galactomannans as natural products in pharmaceutical industries.
Collapse
|
26
|
Calvo-Martín G, Plano D, Encío I, Sanmartín C. Novel N, N'-Disubstituted Selenoureas as Potential Antioxidant and Cytotoxic Agents. Antioxidants (Basel) 2021; 10:antiox10050777. [PMID: 34068900 PMCID: PMC8156206 DOI: 10.3390/antiox10050777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/11/2021] [Indexed: 12/28/2022] Open
Abstract
A series of 30 novel N,N disubstituted selenoureas were synthesized, characterized, and their antioxidant ability was tested using 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2′-azino-bis(3-ethylbenzthiazoline-6-sulfonic acid (ABTS) assays. Additionally, their cytotoxic activity was tested in vitro in a panel of three different cancer (breast, lung and colon) and two normal cell lines. Each selenourea entity contains a para-substituted phenyl ring with different electron-withdrawing and electron-donating groups, and different aliphatic and aromatic nuclei. All of the synthesized selenoureas present antioxidant capacity at high concentrations in the DPPH assay, and three of them (2b, 2c and 2d) showed greater radical scavenging capacity than ascorbic acid at lower concentrations. These results were confirmed by the ABTS assay, where these novel selenoureas present even higher antioxidant capacity than the reference compound Trolox. On the other hand, 10 selenoureas present IC50 values below 10 µM in at least one cancer cell line, resulting in the adamantyl nucleus (6a–6e), the most interesting in terms of activity and selectivity. Outstanding results were found for selenourea 6c, tested in the NCI60 cell line panel and showing an average GI50 of 1.49 µM for the 60 cell lines, and LC50 values ranging from 9.33 µM to 4.27 µM against 10 of these cancer cell lines. To gain insight into its anticancer activity mechanism, we investigated the cell cycle progression of the promising compound 6c, as well as the type of programmed-cell death in a colon cancer cell line it provokes (HT-29). Compound 6c provoked S phase cell cycle arrest and the induction of cell death was independent of caspase activation, suggesting autophagy, though this assertion requires additional studies. Overall, we envision that this compound can be further developed for the potential treatment of colon cancer.
Collapse
Affiliation(s)
- Gorka Calvo-Martín
- Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain; (G.C.-M.); (D.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, E-31008 Pamplona, Spain;
| | - Daniel Plano
- Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain; (G.C.-M.); (D.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, E-31008 Pamplona, Spain;
| | - Ignacio Encío
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, E-31008 Pamplona, Spain;
- Departamento de Ciencias de la Salud, Universidad Pública de Navarra, Avda. Barañain s/n, E-31008 Pamplona, Spain
| | - Carmen Sanmartín
- Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain; (G.C.-M.); (D.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, E-31008 Pamplona, Spain;
- Correspondence: ; Tel.: +34-948425600 (ext. 806388)
| |
Collapse
|
27
|
Gupta DD, Mishra S, Verma SS, Shekher A, Rai V, Awasthee N, Das TJ, Paul D, Das SK, Tag H, Chandra Gupta S, Hui PK. Evaluation of antioxidant, anti-inflammatory and anticancer activities of diosgenin enriched Paris polyphylla rhizome extract of Indian Himalayan landraces. JOURNAL OF ETHNOPHARMACOLOGY 2021; 270:113842. [PMID: 33460752 DOI: 10.1016/j.jep.2021.113842] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/27/2020] [Accepted: 01/08/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional medicinal plants have gained attention as a potential therapeutic agent to combat cancer and inflammation. Diosgenin rich fresh extracts of Paris polyphylla rhizome from Indian Himalaya is traditionally used as wound healing, anti-bleeding, anti-inflammatory and anti-cancer agent by the folk healers. AIM OF THE STUDY Present study was aimed to prepare two types of extracts from Paris polyphylla rhizome of Indian Himalayan landraces - 1. ethanolic extract of Paris polyphylla rhizome (EEPPR) and 2. Diosgenin enriched Paris polyphylla rhizome extract (DPPE), quantification of diosgenin content, and to evaluate their in vitro anti-oxidant, in vivo anti-inflammatory and in vitro cytotoxicity and anti-cancer activities of the DPPE. MATERIALS AND METHODS Diosgenin content of EEPPR was quantified through GC-MS while diosgenin content of DPPE was quantified through HPTLC, and the diosgenin yield from EEPPR and DPPE were compared. In vitro antioxidant activities of DPPE were performed using DPPH, NOD, RP and SOD assay while in vivo anti-inflammatory activity of DPPE were evaluated in dextran induced hind paw edema in rats. In vitro cytotoxicity and anti-cancer activities of DPPE were evaluated in human breast cancer cell lines (MCF-7, MDA-MB-231), cervical cancer cell lines (HeLa) and Hep-2 cell lines. RESULTS EEPPR obtained through cold extraction method using 70% ethanol showed maximum diosgenin content of 17.90% quantified through GC-MS while similar compounds pennogenin (3.29%), 7β-Dehydrodiosgenin (1.90%), 7-Ketodiosgenin acetate (1.14%), and 7 β-hydroxydiosgenin (0.55%) were detected in low concentration, and thus confirmed diosgenin as major and lead phytochemical. However, DPPE obtained through both cold and repeated hot extraction with the same solvent (70% ethanol) showed diosgenin content of 60.29% which is significantly higher (p < 0.001) than the diosgenin content in EEPPR. DPPE demonstrated significant in vitro antioxidant activities by dose-dependently quenched (p < 0.001) SOD free radicals by 76.66%, followed by DPPH (71.43%), NOD (67.35%), and RP (63.74%) at a max concentration of 2 μg/μl of ascorbic acid and test drugs with remarkable IC50 values (p < 0.01). Further, DPPE also showed potent anti-inflammatory activities by dose-dependently suppressed dextran induced paw edema in rats (p < 0.01) from 2 h to 4 h. DPPE suppressed the proliferation of MCF-7, MDA-MB-231, Hep-2 and HeLa cell lines. Maximum activity was observed in MCF-7 cells. The DPPE also induced apoptosis in MCF-7 cell lines as measured by AO/PI and DAPI staining, as well as DNA laddering, cell cycle analysis and phosphatidylserine externalization assay. The growth-inhibitory effect of DPPE on MCF-7 breast cancer cells was further confirmed from the colony-formation assay. DPPE upregulated expression of Bax and downregulated Bcl-2 and survivin mRNA transcripts. CONCLUSION DPPE obtained through both cold and repeated hot extraction using ethanol showed significantly higher content of diosgenin than the diosgenin content detected in EEPPR. However, diosgenin yield of both the extracts (EEPPR & DPPE) clearly confirmed diosgenin as major and lead phytochemical of Paris polyphylla rhizome of Indian Himalayan landraces. Further, DPPE also demonstrated potent in vitro anti-oxidative and in vivo anti-inflammatory activities and showed in vitro cytotoxicity and significant anti-cancer (apoptosis) effects in MCF-7 breast cancer cells.
Collapse
Affiliation(s)
- Debmalya Das Gupta
- Department of Biotechnology, National Institute of Technology (NIT)-Arunachal Pradesh, Yupia, 791112, Papum Pare, Arunachal Pradesh, India.
| | - Shruti Mishra
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Sumit Singh Verma
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Anusmita Shekher
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Vipin Rai
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Nikee Awasthee
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Tridip J Das
- Department of Biotechnology, National Institute of Technology (NIT)-Arunachal Pradesh, Yupia, 791112, Papum Pare, Arunachal Pradesh, India.
| | - Dipayan Paul
- Department of Biotechnology, National Institute of Technology (NIT)-Arunachal Pradesh, Yupia, 791112, Papum Pare, Arunachal Pradesh, India.
| | - Sanjib K Das
- Department of Biotechnology, National Institute of Technology (NIT)-Arunachal Pradesh, Yupia, 791112, Papum Pare, Arunachal Pradesh, India.
| | - Hui Tag
- Pharmacognosy Research Laboratory, Department of Botany, Rajiv Gandhi University, Rono Hills, Doimukh, 791112, Arunachal Pradesh, India.
| | - Subash Chandra Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Pallabi K Hui
- Department of Biotechnology, National Institute of Technology (NIT)-Arunachal Pradesh, Yupia, 791112, Papum Pare, Arunachal Pradesh, India.
| |
Collapse
|
28
|
Xia X, Chen Y, Wang L, Yang ZG, Ma XD, Zhao ZG, Yang HJ. Synthesis of diosgenyl quaternary ammonium derivatives and their antitumor activity. Steroids 2021; 166:108774. [PMID: 33285175 DOI: 10.1016/j.steroids.2020.108774] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/12/2020] [Accepted: 11/30/2020] [Indexed: 01/13/2023]
Abstract
Giosgenin is a naturally steroidal saponin exhibiting a variety of biological activities including antitumor ones. A series of novel diosgenyl quaternary ammonium derivatives were designed and synthesized to develop potential anti-tumor agents in our research. All novel derivatives were characterized by 1H NMR, 13C NMR and HR-MS, and evaluated for their in vitro anti-proliferative activities using MTT assay. The human cancer cell lines were A549 (Human lung cancer cell), H1975 (Human lung adenocarcinoma cell), A431 (Human skin squamous cell carcinoma), HCT-116 (Human colorectal adenocarcinoma cell), Aspc-1 (Human metastatic pancreatic cancer cell), Ramos (Human B lymphoma cell), HBE (Human bronchial epithelioid cell) and LO2 (Human normal hepatocyte).
Collapse
Affiliation(s)
- Xi Xia
- Key Laboratory of Basic Chemistry of the State Ethnic Affairs Commission, School of Chemistry and Environment, Southwest Minzu University, Chengdu 610041, PR China
| | - Yu Chen
- Key Laboratory of Basic Chemistry of the State Ethnic Affairs Commission, School of Chemistry and Environment, Southwest Minzu University, Chengdu 610041, PR China
| | - Lin Wang
- Key Laboratory of Basic Chemistry of the State Ethnic Affairs Commission, School of Chemistry and Environment, Southwest Minzu University, Chengdu 610041, PR China
| | - Zhi-Gang Yang
- Key Laboratory of Basic Chemistry of the State Ethnic Affairs Commission, School of Chemistry and Environment, Southwest Minzu University, Chengdu 610041, PR China
| | - Xiao-Dong Ma
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Zhi-Gang Zhao
- Key Laboratory of Basic Chemistry of the State Ethnic Affairs Commission, School of Chemistry and Environment, Southwest Minzu University, Chengdu 610041, PR China
| | - Hong-Jun Yang
- Key Laboratory of Basic Chemistry of the State Ethnic Affairs Commission, School of Chemistry and Environment, Southwest Minzu University, Chengdu 610041, PR China.
| |
Collapse
|
29
|
Glutathione peroxidase-like functions of 1,2-diselenane-4,5-diol and its amphiphilic derivatives: Switchable catalytic cycles depending on peroxide substrates. Bioorg Med Chem 2021; 29:115866. [DOI: 10.1016/j.bmc.2020.115866] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022]
|
30
|
Abstract
An efficient and practical method for the straightforward construction of unsymmetrical selenoureas and cycloselenoureas via the combination of selenium powder, chloroform, and two different amines was comprehensively achieved in one-pot with only the assistance of a base under mild conditions. Thirty-three new structures of unsymmetrical selenoureas including three chiral examples and eight cycloselenoureas were achieved. 1,1-Dimethyl-3-phenylselenourea II, which shows good fungicidal activity, was practically synthesized through this protocol in gram-scale. Isoselenocyanate was further confirmed as a key intermediate by control experiment.
Collapse
Affiliation(s)
- Lai Li
- College of Chemistry and Materials Engineering, Wenzhou University, Chashan University Town, Wenzhou, Zhejiang Province 325035, People's Republic of China
| | - Jiaqi Wu
- College of Chemistry and Materials Engineering, Wenzhou University, Chashan University Town, Wenzhou, Zhejiang Province 325035, People's Republic of China
| | - Linsha Wei
- College of Chemistry and Materials Engineering, Wenzhou University, Chashan University Town, Wenzhou, Zhejiang Province 325035, People's Republic of China
| | - Jianmei Lu
- College of Chemistry and Materials Engineering, Wenzhou University, Chashan University Town, Wenzhou, Zhejiang Province 325035, People's Republic of China
| | - Xuefeng Jiang
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
| |
Collapse
|
31
|
Begines P, Sevilla-Horrillo L, Puerta A, Puckett R, Bayort S, Lagunes I, Maya I, Padrón JM, López Ó, Fernández-Bolaños JG. Masked Phenolic-Selenium Conjugates: Potent and Selective Antiproliferative Agents Overcoming P-gp Resistance. Pharmaceuticals (Basel) 2020; 13:ph13110358. [PMID: 33142908 PMCID: PMC7692337 DOI: 10.3390/ph13110358] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/23/2020] [Accepted: 10/28/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer accounts for one of the most complex diseases nowadays due to its multifactorial nature. Despite the vast number of cytotoxic agents developed so far, good therapeutic approaches are not always reached. In recent years, multitarget drugs are gaining great attention against multifactorial diseases in contraposition to polypharmacy. Herein we have accomplished the conjugation of phenolic derivatives with an ample number of organochalcogen motifs with the aim of developing novel antiproliferative agents. Their antioxidant, and antiproliferative properties (against six tumour and one non-tumour cell lines) were analysed. Moreover, in order to predict P-gp-mediated chemoresistance, the P-glycoprotein assay was also conducted in order to determine whether compounds prepared herein could behave as substrates of that glycoprotein. Selenium derivatives were found to be significantly stronger antiproliferative agents than their sulfur isosters. Moreover, the length and the nature of the tether, together with the nature of the organoselenium scaffold were also found to be crucial features in the observed bioactivities. The lead compound, bearing a methylenedioxyphenyl moiety, and a diselenide functionality, showed a good activity (GI50 = 0.88‒2.0 µM) and selectivity towards tumour cell lines (selectivity index: 14‒32); moreover, compounds considered herein were not substrates for the P-gp efflux pump, thus avoiding the development of chemoresistance coming from such mechanism, commonly found for widely-used chemotherapeutic agents.
Collapse
Affiliation(s)
- Paloma Begines
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071 Seville, Spain; (P.B.); (S.B.); (I.M.)
| | - Lucía Sevilla-Horrillo
- Escuela Politécnica Superior, Universidad de Sevilla, Virgen de África 7, E-41011 Seville, Spain; (L.S.-H.); (R.P.)
| | - Adrián Puerta
- BioLab, Instituto Universitario de Bio-Orgánica “Antonio González” (IUBO-AG), Universidad de La Laguna, c/ Astrofísico Francisco Sánchez 2, E-38206 La Laguna, Spain; (A.P.); (I.L.)
| | - Rebecca Puckett
- Escuela Politécnica Superior, Universidad de Sevilla, Virgen de África 7, E-41011 Seville, Spain; (L.S.-H.); (R.P.)
| | - Samuel Bayort
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071 Seville, Spain; (P.B.); (S.B.); (I.M.)
| | - Irene Lagunes
- BioLab, Instituto Universitario de Bio-Orgánica “Antonio González” (IUBO-AG), Universidad de La Laguna, c/ Astrofísico Francisco Sánchez 2, E-38206 La Laguna, Spain; (A.P.); (I.L.)
| | - Inés Maya
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071 Seville, Spain; (P.B.); (S.B.); (I.M.)
| | - José M. Padrón
- BioLab, Instituto Universitario de Bio-Orgánica “Antonio González” (IUBO-AG), Universidad de La Laguna, c/ Astrofísico Francisco Sánchez 2, E-38206 La Laguna, Spain; (A.P.); (I.L.)
- Correspondence: (J.M.P.); (Ó.L.); (J.G.F.-B.); Tel.: +34-922-316-502 (J.M.P.) ext. 6126; +34-954-559-997 (Ó.L.); +34-954-550-996 (J.G.F.-B.)
| | - Óscar López
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071 Seville, Spain; (P.B.); (S.B.); (I.M.)
- Correspondence: (J.M.P.); (Ó.L.); (J.G.F.-B.); Tel.: +34-922-316-502 (J.M.P.) ext. 6126; +34-954-559-997 (Ó.L.); +34-954-550-996 (J.G.F.-B.)
| | - José G. Fernández-Bolaños
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071 Seville, Spain; (P.B.); (S.B.); (I.M.)
- Correspondence: (J.M.P.); (Ó.L.); (J.G.F.-B.); Tel.: +34-922-316-502 (J.M.P.) ext. 6126; +34-954-559-997 (Ó.L.); +34-954-550-996 (J.G.F.-B.)
| |
Collapse
|
32
|
Michalak O, Krzeczyński P, Cieślak M, Cmoch P, Cybulski M, Królewska-Golińska K, Kaźmierczak-Barańska J, Trzaskowski B, Ostrowska K. Synthesis and anti-tumour, immunomodulating activity of diosgenin and tigogenin conjugates. J Steroid Biochem Mol Biol 2020; 198:105573. [PMID: 32017993 DOI: 10.1016/j.jsbmb.2019.105573] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022]
Abstract
A series of novel diosgenin (DSG) and tigogenin (TGG) derivatives with diosgenin or tigogenin steroid aglycons linked to levulinic and 3,4-dihydroxycinnamic acids, dipeptides and various amino acids by an ester bond at the C3-oxygen atom of the steroid skeleton has been synthesized. Diosgenyl esters have been prepared by an esterification reaction (DCC/DMAP) of diosgenin with the corresponding acids. All analogues have been evaluated in vitro for their antiproliferative profile against cancer cell lines (MCF-7, MDA-MB-231, PC-3) and human umbilical vein endothelial cells (HUVEC). Analogue2c (l-serine derivative of TGG), the best representative of the series showed IC50 of 1.5 μM (MCF-7), and induced apoptosis in MCF-7 by activating caspase-3/7. The immunomodulatory properties of six synthesized analogues have been determined by examining their effects on the expression of cytokine genes essential for the functioning of the human immune system (IL-1, IL-4, IL-10, IL-12 and TNF-α). Biological evaluation has revealed that new compounds 4c and 16a do not induce the expression of pro-inflammatory cytokines in THP-1 cells after the lipopolysaccharide (LPS) stimulation. They also stimulate the expression of anti-inflammatory IL-10 that acts stronger than diosgenin itself. An in silico ADME properties(absorption, distribution, metabolism, excretion) study was also performed to predict the pharmacokinetic profile of the synthesized compounds. To shed light on the molecular interactions between the synthesized compounds and the glucocorticoid receptor and the estrogen receptor, 2c, 4c and 16a compounds were docked into the active binding sites of these receptors. The in silico and in vitro data suggested that this new group of compounds might be considered as a promising scaffold for further modification of more potent and selective anticancer and immunomodulatory agents.
Collapse
Affiliation(s)
- O Michalak
- Łukasiewicz Research Network-Pharmaceutical Research Institute, 8 Rydygiera Str., 01-793 Warsaw, Poland.
| | - P Krzeczyński
- Łukasiewicz Research Network-Pharmaceutical Research Institute, 8 Rydygiera Str., 01-793 Warsaw, Poland
| | - M Cieślak
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 112 Sienkiewicza Str., 90-363 Łódź, Poland
| | - P Cmoch
- Institute of Organic Chemistry, Polish Academy of Sciences, 44/52 Kasprzaka Str., 01-224 Warsaw, Poland
| | - M Cybulski
- Łukasiewicz Research Network-Pharmaceutical Research Institute, 8 Rydygiera Str., 01-793 Warsaw, Poland
| | - K Królewska-Golińska
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 112 Sienkiewicza Str., 90-363 Łódź, Poland
| | - J Kaźmierczak-Barańska
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 112 Sienkiewicza Str., 90-363 Łódź, Poland
| | - B Trzaskowski
- Chemical and Biological Systems Simulation Lab, Center of New Technologies, University of Warsaw, 2C Banacha Str., 02-097 Warsaw, Poland
| | - K Ostrowska
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| |
Collapse
|
33
|
Spectral and computational studies on regioselective synthesis of 4-oxo-6-phenyl-2-selenoxo-1,2,3,4-tetrahydropyrimidine-5-carbonitrile. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2019.127408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
34
|
Ruberte AC, Ramos-Inza S, Aydillo C, Talavera I, Encío I, Plano D, Sanmartín C. Novel N, N'-Disubstituted Acylselenoureas as Potential Antioxidant and Cytotoxic Agents. Antioxidants (Basel) 2020; 9:antiox9010055. [PMID: 31936213 PMCID: PMC7023466 DOI: 10.3390/antiox9010055] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/30/2019] [Accepted: 01/06/2020] [Indexed: 12/23/2022] Open
Abstract
Selenium compounds are pivotal in medicinal chemistry for their antitumoral and antioxidant properties. Forty seven acylselenoureas have been designed and synthesized following a fragment-based approach. Different scaffolds, including carbo- and hetero-cycles, along with mono- and bi-cyclic moieties, have been linked to the selenium containing skeleton. The dose- and time-dependent radical scavenging activity for all of the compounds were assessed using the in vitro 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2′-azino-bis(3-ethylbenzthiazoline-6-sulfonic acid) (ABTS) assays. Some of them showed a greater radical scavenging capacity at low doses and shorter times than ascorbic acid. Therefore, four compounds were evaluated to test their protective effects against H2O2-induced oxidative stress. One derivative protected cells against H2O2-induced damage, increasing cell survival by up to 3.6-fold. Additionally, in vitro cytotoxic activity of all compounds was screened against several cancer cells. Eight compounds were selected to determine their half maximal inhibitory concentration (IC50) values towards breast and lung cancer cells, along with their selectivity indexes. The breast cancer cells turned out to be much more sensitive than the lung. Two compounds (5d and 10a) stood out with IC50 values between 4.2 μM and 8.0 μM towards MCF-7 and T47D cells, with selectivity indexes greater than 22.9. In addition, compound 10b exhibited dual antioxidant and cytotoxic activities. Although further evidence is needed, the acylselenourea scaffold could be a feasible frame to develop new dual agents.
Collapse
Affiliation(s)
- Ana Carolina Ruberte
- Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain; (A.C.R.); (S.R.-I.); (C.A.); (I.T.); (D.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, 31008 Pamplona, Spain;
| | - Sandra Ramos-Inza
- Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain; (A.C.R.); (S.R.-I.); (C.A.); (I.T.); (D.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, 31008 Pamplona, Spain;
| | - Carlos Aydillo
- Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain; (A.C.R.); (S.R.-I.); (C.A.); (I.T.); (D.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, 31008 Pamplona, Spain;
| | - Irene Talavera
- Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain; (A.C.R.); (S.R.-I.); (C.A.); (I.T.); (D.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, 31008 Pamplona, Spain;
| | - Ignacio Encío
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, 31008 Pamplona, Spain;
- Departamento de Ciencias de la Salud, Universidad Pública de Navarra, Avda. Barañain s/n, 31008 Pamplona, Spain
| | - Daniel Plano
- Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain; (A.C.R.); (S.R.-I.); (C.A.); (I.T.); (D.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, 31008 Pamplona, Spain;
| | - Carmen Sanmartín
- Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain; (A.C.R.); (S.R.-I.); (C.A.); (I.T.); (D.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, 31008 Pamplona, Spain;
- Correspondence: ; Tel.: +34-948425600 (ext. 806388)
| |
Collapse
|
35
|
E. Ali T, A. Assiri M, M. Ali M, E. M. Ali A, S. Yahia I, Y. Zahran H. Efficient Synthesis and Anticancer Activities of Some Novel Functionalized (4-Oxo-4H-chromen-3-yl)-2-selenoxo-1,2-dihydropyrimidines. HETEROCYCLES 2020. [DOI: 10.3987/com-20-14324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
36
|
Cui J, Wei M, Pang L, Gan C, Xiao J, Shi H, Zhan J, Liu Z, Huang Y. Synthesis and antiproliferative evaluation of novel steroid-benzisoselenazolone hybrids. Steroids 2019; 152:108502. [PMID: 31545961 DOI: 10.1016/j.steroids.2019.108502] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/31/2019] [Accepted: 09/17/2019] [Indexed: 10/26/2022]
Abstract
The two different types of steroidal benzisoselenazolone hybrids were synthesized by incorporating benzisoselenazolone scaffold into dehydroepiandrosterone and B-norcholesterol. The antiproliferative activity of the synthesized compounds against some carcinoma cell lines were investigated. The results showed that some of these compounds have better inhibitory activity than abiraterone on the proliferation of tumor cells associated with human growth hormone, and have less cytotoxicity on normal human cells. In particular, the IC50 values of the compound 8a and 8f are 5.4 and 6.5 µmol/L against human ovarian carcinoma (SKOV3) cell line, and possess SI values of 13.9 and 10.5, respectively. The information obtained from the studies may be useful for the design of novel chemotherapeutic drugs.
Collapse
Affiliation(s)
- Jianguo Cui
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China; Guangxi Colleges and University Key Laboratory of Beibu Gulf Oil and Natural Gas Resource Effective Utilization, Beibuwan University, China.
| | - Meizhen Wei
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China
| | - Liping Pang
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China
| | - Chunfang Gan
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China
| | - Junan Xiao
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China
| | - Haixin Shi
- Guangxi Colleges and University Key Laboratory of Beibu Gulf Oil and Natural Gas Resource Effective Utilization, Beibuwan University, China
| | - Junyan Zhan
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China
| | - Zhiping Liu
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China
| | - Yanmin Huang
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, PR China.
| |
Collapse
|
37
|
Fouda AM, Assiri MA, Ali TE. Facile synthesis of some new functionalized 2-selenoxopyrimidines. PHOSPHORUS SULFUR 2019. [DOI: 10.1080/10426507.2019.1694023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Ahmed M. Fouda
- Department of Chemistry, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | - Mohammed A. Assiri
- Department of Chemistry, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | - Tarik E. Ali
- Department of Chemistry, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
38
|
Mironov ME, Oleshko OS, Pokrovskii MA, Rybalova TV, Pechurov VK, Pokrovskii AG, Cheresis SV, Mishinov SV, Stupak VV, Shults EE. 6-(4'-Aryl-1',2',3'-triazolyl)-spirostan-3,5-diols and 6-(4'-Aryl-1',2',3'-triazolyl)-7-hydroxyspirosta-1,4-dien-3-ones: Synthesis and analysis of their cytotoxicity. Steroids 2019; 151:108460. [PMID: 31344410 DOI: 10.1016/j.steroids.2019.108460] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 07/10/2019] [Accepted: 07/16/2019] [Indexed: 12/30/2022]
Abstract
In an endeavour to develop potent anti-tumor agents from diosgenin, a series of C-6 derived 1,2,3-triazolyl derivatives were designed and synthesized by employing Cu(I) catalyzed Huisgen 1,3-dipolar cycloaddition reaction of novel azides - (22R,25R)-6β-azidospirostan-3β,5α-diol and 6β-azido-7α-hydroxyspirosta-1,4-dien-3-one with aryl(hetaryl)alkynes. All the derivatives were evaluated for cytotoxic activity by MTT assay against eight different human cancer cell lines: T-cellular leucosis (CEM-13), human monocytes (U-937), breast (MDA-MB-231, BT-474), prostate (DU-145) and glioblastoma (U-87MG, SNB-19, T98G). The results of this study suggested that 6-(4'-aryl-1',2',3'-triazolyl)spirostan-3,5-diols 2, 3, 4, 5 and 6 possessed a promising cytotoxic potential. The corresponding 6-substituted 7-hydroxy-1,4-spirostadien-3-ones shown less cytotoxity on the human cancer cells. Compounds 2, 3, 4, and 5 which demonstrated high grown inhibition against glioma cancer cells U-87 and T98G, and also on the human-derived N118669 primary glioblastoma cell line (with GI50 values in the range of 5-9 μM), were not affected the growth of SNB-19 cells. The data revealed that phenyl, 4-methoxyphenyl, 4-fluorophenyl, 3,4,5-trimethoxyphenyl or 2-pyridinyl substituent in the triazole moiety at the C-6 position significantly improved the anti-tumor activity. The mentioned position at the spirostan core may be favourable for the synthesis of potent anticancer leads from diosgenin.
Collapse
Affiliation(s)
- Maxim E Mironov
- Laboratory of Medicinal Chemistry, Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Lavrentyev Ave, 9, 630090 Novosibirsk, Russian Federation; Novosibirsk State University, Pirogova Str. 2, 630090 Novosibirsk, Russian Federation
| | - Olga S Oleshko
- Novosibirsk State University, Pirogova Str. 2, 630090 Novosibirsk, Russian Federation
| | - Mikhail A Pokrovskii
- Novosibirsk State University, Pirogova Str. 2, 630090 Novosibirsk, Russian Federation
| | - Tatyana V Rybalova
- Laboratory of Medicinal Chemistry, Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Lavrentyev Ave, 9, 630090 Novosibirsk, Russian Federation; Novosibirsk State University, Pirogova Str. 2, 630090 Novosibirsk, Russian Federation
| | - Vladislav K Pechurov
- Novosibirsk State University, Pirogova Str. 2, 630090 Novosibirsk, Russian Federation
| | - Andrey G Pokrovskii
- Novosibirsk State University, Pirogova Str. 2, 630090 Novosibirsk, Russian Federation
| | - Sergey V Cheresis
- Novosibirsk State University, Pirogova Str. 2, 630090 Novosibirsk, Russian Federation
| | - Sergey V Mishinov
- First Department of Neurosurgery, Ya. L. Tsivian Novosibirsk Research Institute of Traumatology and Orthopaedics, Frunze Str. 17, 630091 Novosibirsk, Russian Federation
| | - Vyacheslav V Stupak
- First Department of Neurosurgery, Ya. L. Tsivian Novosibirsk Research Institute of Traumatology and Orthopaedics, Frunze Str. 17, 630091 Novosibirsk, Russian Federation
| | - Elvira E Shults
- Laboratory of Medicinal Chemistry, Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Lavrentyev Ave, 9, 630090 Novosibirsk, Russian Federation; Novosibirsk State University, Pirogova Str. 2, 630090 Novosibirsk, Russian Federation.
| |
Collapse
|
39
|
Andreeva OV, Belenok MG, Saifina LF, Shulaeva MM, Dobrynin AB, Sharipova RR, Voloshina AD, Saifina AF, Gubaidullin AT, Khairutdinov BI, Zuev YF, Semenov VE, Kataev VE. Synthesis of novel 1,2,3-triazolyl nucleoside analogues bearing uracil, 6-methyluracil, 3,6-dimethyluracil, thymine, and quinazoline-2,4-dione moieties. Tetrahedron Lett 2019. [DOI: 10.1016/j.tetlet.2019.151276] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
40
|
Lagunes I, Begines P, Silva A, Galán AR, Puerta A, Fernandes MX, Maya I, Fernández-Bolaños JG, López Ó, Padrón JM. Selenocoumarins as new multitarget antiproliferative agents: Synthesis, biological evaluation and in silico calculations. Eur J Med Chem 2019; 179:493-501. [PMID: 31271961 DOI: 10.1016/j.ejmech.2019.06.073] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 05/28/2019] [Accepted: 06/26/2019] [Indexed: 11/26/2022]
Abstract
Herein we report a straightforward preparation of new antiproliferative agents based on the hybridization of a coumarin skeleton and an organoselenium motif. Three families were obtained: isoselenocyanate, selenocarbamates and selenoureas. The main purpose of these hybrid structures is the development of new antiproliferative agents with a multitarget mode of action. A strong correlation between the nature of the organosenium scaffold and the antiproliferative activity was observed. Thus, whereas selenocarbamates proved to be inactive, or moderate antiproliferative agents, isoselenocyanate and most of the selenoureas behaved as strong antiproliferative agents, with GI50 values within the low micromolar range. Interestingly, a good selectivity toward tumor cell lines was found for some of the compounds. Moreover, an increase in the ROS level was observed for tumor cells, and accordingly, these pro-oxidant species might be involved in their mode of action. Overall, title compounds were found not to be substrates for P-glycoprotein, which is overexpressed in many cancer cells as a way of detoxification, and thus, to develop drug resistance. In silico calculations revealed that the selenoderivatives prepared herein might undergo a strong interaction with the active site of HDAC8, and therefore, be potential inhibitors of histone deacetylase 8. In vitro assessment against HDAC8 revealed a strong inhibition of such enzyme exerted by selenoureas, particularly by symmetrical coumarin-containing selenourea. Two compounds showed good antiproliferative data and appear as plausible leads for further testings. The symmetrical coumarin 6 displays the best in vitro inhibition of HDAC8, but is affected by P-gp. In contrast, the N-butyl selenourea coumarin derivative 5a escapes P-gp resistance but has lower HDAC8 inhibition activity.
Collapse
Affiliation(s)
- Irene Lagunes
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Centro de Investigaciones Biomédicas de Canarias (CIBICAN), Universidad de La Laguna, Apartado 456, E-38071, La Laguna, Spain
| | - Paloma Begines
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071, Sevilla, Spain
| | - Adrián Silva
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071, Sevilla, Spain
| | - Alexis R Galán
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Centro de Investigaciones Biomédicas de Canarias (CIBICAN), Universidad de La Laguna, Apartado 456, E-38071, La Laguna, Spain
| | - Adrián Puerta
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Centro de Investigaciones Biomédicas de Canarias (CIBICAN), Universidad de La Laguna, Apartado 456, E-38071, La Laguna, Spain
| | - Miguel X Fernandes
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Centro de Investigaciones Biomédicas de Canarias (CIBICAN), Universidad de La Laguna, Apartado 456, E-38071, La Laguna, Spain
| | - Inés Maya
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071, Sevilla, Spain
| | - José G Fernández-Bolaños
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071, Sevilla, Spain.
| | - Óscar López
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071, Sevilla, Spain.
| | - José M Padrón
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Centro de Investigaciones Biomédicas de Canarias (CIBICAN), Universidad de La Laguna, Apartado 456, E-38071, La Laguna, Spain.
| |
Collapse
|
41
|
Jastrzebska I, Mellea S, Salerno V, Grzes PA, Siergiejczyk L, Niemirowicz-Laskowska K, Bucki R, Monti B, Santi C. PhSeZnCl in the Synthesis of Steroidal β-Hydroxy-Phenylselenides Having Antibacterial Activity. Int J Mol Sci 2019; 20:ijms20092121. [PMID: 31032813 PMCID: PMC6539910 DOI: 10.3390/ijms20092121] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 01/07/2023] Open
Abstract
We report here the reaction of in situ prepared PhSeZnCl with steroid derivatives having an epoxide as an electrophilic functionalization. The corresponding ring-opening reaction resulted to be regio- and stereoselective affording to novel phenylselenium-substituted steroids. Assessment of their antibacterial properties against multidrug-resistant bacteria, such as Pseudomonas aeruginosa Xen 5 strain, indicates an interesting bactericidal activity and their ability to prevent bacterial biofilm formation.
Collapse
Affiliation(s)
- Izabella Jastrzebska
- Institute of Chemistry, University of Białystok, ul. Ciołkowskiego 1K, 15-245 Białystok, Poland.
| | - Stefano Mellea
- Institute of Chemistry, University of Białystok, ul. Ciołkowskiego 1K, 15-245 Białystok, Poland.
| | - Valerio Salerno
- Institute of Chemistry, University of Białystok, ul. Ciołkowskiego 1K, 15-245 Białystok, Poland.
| | - Pawel Adam Grzes
- Institute of Chemistry, University of Białystok, ul. Ciołkowskiego 1K, 15-245 Białystok, Poland.
| | - Leszek Siergiejczyk
- Institute of Chemistry, University of Białystok, ul. Ciołkowskiego 1K, 15-245 Białystok, Poland.
| | - Katarzyna Niemirowicz-Laskowska
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Białystok, ul. Mickiewicza 2C, 15-222 Bialystok, Poland.
| | - Robert Bucki
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Białystok, ul. Mickiewicza 2C, 15-222 Bialystok, Poland.
| | - Bonifacio Monti
- Group of Catalysis and Organic Green Chemistry⁻Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, 06132 Perugia, Italy.
| | - Claudio Santi
- Group of Catalysis and Organic Green Chemistry⁻Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, 06132 Perugia, Italy.
| |
Collapse
|
42
|
Cui J, Pang L, Wei M, Gan C, Liu D, Yuan H, Huang Y. Synthesis and antiproliferative activity of 17-[1',2',3']-selenadiazolylpregnenolone compounds. Steroids 2018; 140:151-158. [PMID: 30296550 DOI: 10.1016/j.steroids.2018.10.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 09/25/2018] [Accepted: 10/01/2018] [Indexed: 12/31/2022]
Abstract
Using pregnenolone as a starting material, some 3-substituted 17-[1',2',3']-selenadiazolylpregnenolone derivatives were synthesized, and their structures were characterized by IR, NMR and HRMS. The in vitro antitumor activity of the compounds was assayed against PC-3、SKOV3、T47D、MCF-7 and HEK293T cell lines. The results show that some compounds display selective antiproliferative activity against PC-3 and SKOV3 cells lines and are almost inactive to normal kidney epithelial cells (HEK293T). The IC50 value are much better than that of abiraterone (positive control).
Collapse
Affiliation(s)
- Jianguo Cui
- College of Chemistry and Material Science, Guangxi Teachers Education University, Nanning 530001, PR China; College of Petroleum and Chemical Engineering, Qizhou University, Qizhou, PR China
| | - Liping Pang
- College of Chemistry and Material Science, Guangxi Teachers Education University, Nanning 530001, PR China
| | - Meizhen Wei
- College of Chemistry and Material Science, Guangxi Teachers Education University, Nanning 530001, PR China
| | - Chunfang Gan
- College of Chemistry and Material Science, Guangxi Teachers Education University, Nanning 530001, PR China
| | - Dandan Liu
- College of Chemistry and Material Science, Guangxi Teachers Education University, Nanning 530001, PR China
| | - Haiyan Yuan
- College of Chemistry and Material Science, Guangxi Teachers Education University, Nanning 530001, PR China
| | - Yanmin Huang
- College of Chemistry and Material Science, Guangxi Teachers Education University, Nanning 530001, PR China.
| |
Collapse
|
43
|
Barbosa FAR, Siminski T, Canto RFS, Almeida GM, Mota NSRS, Ourique F, Pedrosa RC, Braga AL. Novel pyrimidinic selenourea induces DNA damage, cell cycle arrest, and apoptosis in human breast carcinoma. Eur J Med Chem 2018; 155:503-515. [PMID: 29908443 DOI: 10.1016/j.ejmech.2018.06.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 06/07/2018] [Accepted: 06/09/2018] [Indexed: 11/30/2022]
Abstract
Novel pyrimidinic selenoureas were synthesized and evaluated against tumour and normal cell lines. Among these, the compound named 3j initially showed relevant cytotoxicity and selectivity for tumour cells. Three analogues of 3j were designed and synthesized keeping in view the structural requirements of this compound. Almost all the tested compounds displayed considerable cytotoxicity. However, 8a, one of the 3j analogues, was shown to be highly selective and cytotoxic, especially for breast carcinoma cells (MCF-7) (IC50 = 3.9 μM). Furthermore, 8a caused DNA damage, inhibited cell proliferation, was able to arrest cell cycle in S phase, and induced cell death by apoptosis in human breast carcinoma cells. Moreover, predictions of pharmacokinetic properties showed that 8a may present good absorption and permeation characteristics for oral administration. Overall, the current study established 8a as a potential drug prototype to be employed as a DNA interactive cytotoxic agent for the treatment of breast cancer.
Collapse
Affiliation(s)
- Flavio A R Barbosa
- Laboratório de Síntese de Substâncias de Selênio Bioativas (LabSelen), Departamento de Química, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Tâmila Siminski
- Laboratório de Bioquímica Experimental (LABIOEX), Departamento de Bioquímica, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Rômulo F S Canto
- Laboratório de Química Medicinal de Compostos de Selênio (QMCSe), Programa de pós-graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Gabriela M Almeida
- Laboratório de Bioquímica Experimental (LABIOEX), Departamento de Bioquímica, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Nádia S R S Mota
- Laboratório de Bioquímica Experimental (LABIOEX), Departamento de Bioquímica, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Fabiana Ourique
- Laboratório de Bioquímica Experimental (LABIOEX), Departamento de Bioquímica, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Rozangela Curi Pedrosa
- Laboratório de Bioquímica Experimental (LABIOEX), Departamento de Bioquímica, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil.
| | - Antonio Luiz Braga
- Laboratório de Síntese de Substâncias de Selênio Bioativas (LabSelen), Departamento de Química, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil.
| |
Collapse
|
44
|
Sethi G, Shanmugam MK, Warrier S, Merarchi M, Arfuso F, Kumar AP, Bishayee A. Pro-Apoptotic and Anti-Cancer Properties of Diosgenin: A Comprehensive and Critical Review. Nutrients 2018; 10:nu10050645. [PMID: 29783752 PMCID: PMC5986524 DOI: 10.3390/nu10050645] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 05/04/2018] [Accepted: 05/16/2018] [Indexed: 12/15/2022] Open
Abstract
Novel and alternative options are being adopted to combat the initiation and progression of human cancers. One of the approaches is the use of molecules isolated from traditional medicinal herbs, edible dietary plants and seeds that play a pivotal role in the prevention/treatment of cancer, either alone or in combination with existing chemotherapeutic agents. Compounds that modulate these oncogenic processes are potential candidates for cancer therapy and may eventually make it to clinical applications. Diosgenin is a naturally occurring steroidal sapogenin and is one of the major bioactive compounds found in dietary fenugreek (Trigonella foenum-graecum) seeds. In addition to being a lactation aid, diosgenin has been shown to be hypocholesterolemic, gastro- and hepato-protective, anti-oxidant, anti-inflammatory, anti-diabetic, and anti-cancer. Diosgenin has a unique structural similarity to estrogen. Several preclinical studies have reported on the pro-apoptotic and anti-cancer properties of diosgenin against a variety of cancers, both in in vitro and in vivo. Diosgenin has also been reported to reverse multi-drug resistance in cancer cells and sensitize cancer cells to standard chemotherapy. Remarkably, diosgenin has also been reported to be used by pharmaceutical companies to synthesize steroidal drugs. Several novel diosgenin analogs and nano-formulations have been synthesized with improved anti-cancer efficacy and pharmacokinetic profile. In this review we discuss in detail the multifaceted anti-cancer properties of diosgenin that have found application in pharmaceutical, functional food, and cosmetic industries; and the various intracellular molecular targets modulated by diosgenin that abrogate the oncogenic process.
Collapse
Affiliation(s)
- Gautam Sethi
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal University, Bangalore 560065, India.
| | - Myriam Merarchi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, 18301 N. Miami Avenue, Miami, FL 33169, USA.
| |
Collapse
|
45
|
Garnica P, Encío I, Plano D, Palop JA, Sanmartín C. Combined Acylselenourea-Diselenide Structures: New Potent and Selective Antitumoral Agents as Autophagy Activators. ACS Med Chem Lett 2018; 9:306-311. [PMID: 29670691 DOI: 10.1021/acsmedchemlett.7b00482] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 03/13/2018] [Indexed: 12/19/2022] Open
Abstract
A series of 16 new diselenide-acylselenourea conjugates have been designed following the fragment-based drug strategy. Compound in vitro cytotoxic potential was evaluated against six human cancer cell lines and two nonmalignant derived cell lines with the aim of determining their potency and selectivity. Nine derivatives exhibited GI50 values under 10 μM in at least four cancer cell lines. A clear gap situated phenyl substitution over heterocyclic moieties in terms of selectivity. Among carbocyclic compounds, derivatives 2 and 7 significantly inhibited cell growth of breast adenocarcinoma cells with GI50 values of 1.30 and 0.15 nM, respectively, with selectivity indexes 12 and 121 times higher than those obtained for doxorubicin. Preliminary mechanistic studies indicated that compounds 2 and 7 induce cell cycle arrest and autophagy-dependent cell death evidenced by the blockage of cell death with pretreatment with wortmannin or chloroquine and confirmed by the upregulation of the markers Beclin1 and LC3B in MCF-7 cells.
Collapse
Affiliation(s)
- Pablo Garnica
- University of Navarra, Faculty of Pharmacy and Nutrition, Department of Organic and Pharmaceutical Chemistry, Irunlarrea 1, E-31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| | - Ignacio Encío
- Department of Health Sciences, Public University of Navarra, Avda. Barañain s/n, E-31008 Pamplona, Spain
| | - Daniel Plano
- University of Navarra, Faculty of Pharmacy and Nutrition, Department of Organic and Pharmaceutical Chemistry, Irunlarrea 1, E-31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| | - Juan A. Palop
- University of Navarra, Faculty of Pharmacy and Nutrition, Department of Organic and Pharmaceutical Chemistry, Irunlarrea 1, E-31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| | - Carmen Sanmartín
- University of Navarra, Faculty of Pharmacy and Nutrition, Department of Organic and Pharmaceutical Chemistry, Irunlarrea 1, E-31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| |
Collapse
|
46
|
Fuentes-Aguilar A, Romero-Hernández LL, Arenas-González A, Merino-Montiel P, Montiel-Smith S, Meza-Reyes S, Vega-Báez JL, Plata GB, Padrón JM, López Ó, Fernández-Bolaños JG. New selenosteroids as antiproliferative agents. Org Biomol Chem 2018; 15:5041-5054. [PMID: 28574071 DOI: 10.1039/c7ob00458c] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Starting from natural steroids (diosgenin, hecogenin, smilagenin, estrone), we have prepared a wide panel of selenoderivatives, including benzoselenazolones, selenosemicarbazones, isoselenocyanates, selenoureas, selenocyanates and diselenides, with the aim of developing new families of potential chemotherapeutic agents. The modification of the organoselenium moieties, and their position on the steroid provided valuable information concerning the antiproliferative activities. Among all the families accessed herein, the best profile was achieved for selenoureas on the A ring of estrone, which exhibited GI50 values in the range 2.0-4.1 μM for all the tested tumor cell lines, with increased potency compared with commonly used chemotherapeutic agents, like 5-fluorouracil and cisplatin. Cell cycle analysis revealed that selenoureas induced accumulation of cells in the G1 phase of the cell cycle in the breast cancer cell lines HBL-100 and T-47D; therefore, a different mechanism than cisplatin, that induces cell cycle accumulation in the S phase as a result of DNA damage, must be involved. In the rest of the tumor cells, a slight increase of the S compartment was observed. Moreover, selenosteoids turned out to be excellent glutathione peroxidase (GPx) mimics for the catalytic removal of deleterious H2O2 (t1/2 8.0-22.5 min) and alkyl peroxides (t1/2 23.0-38.9 min) when used in substoichiometric amounts (1% molar ratio), thus providing a valuable tool for reducing the intrinsic oxidative stress in tumor progression.
Collapse
Affiliation(s)
- Alma Fuentes-Aguilar
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, PUE, Mexico.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Sidoryk K, Rárová L, Oklešťková J, Pakulski Z, Strnad M, Cmoch P, Luboradzki R. Synthesis of 28a-homoselenolupanes and 28a-homoselenolupane saponins. Org Biomol Chem 2018; 14:10238-10248. [PMID: 27735956 DOI: 10.1039/c6ob01938b] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
A practical synthesis of 28a-homo-28a-selenolupane triterpenes and the corresponding selenosaponins containing d-mannose, l-arabinose, l-rhamnose, and d-idose moieties is described. Selenium containing triterpenes were obtained from the readily available 3-O-allyl-homobetulin mesylate by nucleophilic substitution with the selenocyanate ion which upon reduction of the -SeCN group afforded the free selenol. Glycosylation using classical Schmidt donors gave 1,2-trans selenosaponins as the main product as well as minute amounts of 1,2-cis isomers. This is one of the very few examples of the synthesis of selenoglycosides by direct glycosylation of free selenols. The studied selenol showed high resistance to air oxidation resulting in good stability during the synthesis of selenolupane derivatives. Cytotoxic activities of new homoselenolupane derivatives were also evaluated in vitro and revealed that some triterpenes exhibited an interesting profile against human cancer cell lines.
Collapse
Affiliation(s)
- Katarzyna Sidoryk
- Institute of Organic Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland. and Pharmaceutical Research Institute, Rydygiera 8, 01-793 Warsaw, Poland
| | - Lucie Rárová
- Department of Chemical Biology and Genetics, Centre of the Region Haná for Biotechnological and Agricultural Research, Palacký University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic
| | - Jana Oklešťková
- Laboratory of Growth Regulators, Centre of the Region Haná for Biotechnological and Agricultural Research, Institute of Experimental Botany ASCR & Palacký University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic.
| | - Zbigniew Pakulski
- Institute of Organic Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland.
| | - Miroslav Strnad
- Laboratory of Growth Regulators, Centre of the Region Haná for Biotechnological and Agricultural Research, Institute of Experimental Botany ASCR & Palacký University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic.
| | - Piotr Cmoch
- Institute of Organic Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland.
| | - Roman Luboradzki
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| |
Collapse
|
48
|
Abstract
Aim: The increasing number of cancer cases has stimulated researchers to seek for novel approaches. We have combined two bioactive moieties: a polyphenolic scaffold and an organoselenium motif. Four different families (isothiocyanates/thioureas, and their selenium isosters) derived from dopamine, (±)-norepinephrine and R-epinephrine were accessed. Results: Heterocumulenes derived from dopamine and β-O-methylnoradrenaline were strong antiproliferative agents (GI50<10 μM). Selenoureas derived from β-O-methylnoradrenaline bearing electron-withdrawing groups (halogen, -NO2, -Ph) on the phenyl ring, were also strong antiproliferative agents, besides exhibiting good antiradical and glutathione peroxidase-like activities. Up to a 14-fold increased activity was achieved compared with classical chemotherapeutic agents, exhibiting also different mechanisms of action (cell cycle assays). Redox analysis on HeLa cells suggested an increase of ROS levels after the incubation period. Conclusion: the combination of organoselenium and phenolic moieties might provide valuable lead compounds with relevant antiproliferative properties.
Collapse
|
49
|
Rocas P, Fernández Y, García-Aranda N, Foradada L, Calvo P, Avilés P, Guillén MJ, Schwartz S, Rocas J, Albericio F, Abasolo I. Improved pharmacokinetic profile of lipophilic anti-cancer drugs using ανβ3-targeted polyurethane-polyurea nanoparticles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 14:257-267. [PMID: 29127040 DOI: 10.1016/j.nano.2017.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 09/04/2017] [Accepted: 10/20/2017] [Indexed: 02/06/2023]
Abstract
Glutathione degradable polyurethane-polyurea nanoparticles (PUUa NP) with a disulfide-rich multiwalled structure and a cyclic RGD peptide as a targeting moiety were synthesized, incorporating a very lipophilic chemotherapeutic drug named Plitidepsin. In vitro studies indicated that encapsulated drug maintained and even improved its cytotoxic activity while in vivo toxicity studies revealed that the maximum tolerated dose (MTD) of Plitidepsin could be increased three-fold after encapsulation. We also found that pharmacokinetic parameters such as maximum concentration (Cmax), area under the curve (AUC) and plasma half-life were significantly improved for Plitidepsin loaded in PUUa NP. Moreover, biodistribution assays in mice showed that RGD-decorated PUUa NP accumulate less in spleen and liver than non-targeted conjugates, suggesting that RGD-decorated nanoparticles avoid sequestration by macrophages from the reticuloendothelial system. Overall, our results indicate that polyurethane-polyurea nanoparticles represent a very valuable nanoplatform for the delivery of lipophilic drugs by improving their toxicological, pharmacokinetic and whole-body biodistribution profiles.
Collapse
Affiliation(s)
- Pau Rocas
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Nanobiotechnological Polymers Division, Ecopol Tech S.L., L'Arboç, Spain
| | - Yolanda Fernández
- Functional Validation & Preclinical Research (FVPR), Drug Delivery and Targeting Group, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - Natalia García-Aranda
- Functional Validation & Preclinical Research (FVPR), Drug Delivery and Targeting Group, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - Laia Foradada
- Functional Validation & Preclinical Research (FVPR), Drug Delivery and Targeting Group, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - Pilar Calvo
- PharmaMar S.A., Colmenar Viejo, Madrid, Spain
| | | | | | - Simó Schwartz
- Functional Validation & Preclinical Research (FVPR), Drug Delivery and Targeting Group, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - Josep Rocas
- Nanobiotechnological Polymers Division, Ecopol Tech S.L., L'Arboç, Spain
| | - Fernando Albericio
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain; Department of Organic Chemistry, University of Barcelona, Barcelona, Spain; School of Chemistry & Physics, University of Kwazulu-Natal, Durban, South Africa
| | - Ibane Abasolo
- Functional Validation & Preclinical Research (FVPR), Drug Delivery and Targeting Group, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain.
| |
Collapse
|
50
|
Romero-Hernández LL, Merino-Montiel P, Meza-Reyes S, Vega-Baez JL, López Ó, Padrón JM, Montiel-Smith S. Synthesis of unprecedented steroidal spiro heterocycles as potential antiproliferative drugs. Eur J Med Chem 2017; 143:21-32. [PMID: 29172080 DOI: 10.1016/j.ejmech.2017.10.063] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/09/2017] [Accepted: 10/22/2017] [Indexed: 01/14/2023]
Abstract
Herein we report the straightforward preparation of novel conformationally-restricted steroids from trans-androsterone and estrone, decorated with spiranic oxazolidin-2-one or 2-aminooxazoline motifs at C-17 as potential antiproliferative agents. Such unprecedented pharmacophores were accessed using an aminomethylalcohol derivative at C-17 as the key intermediate; reaction of such functionality with triphosgene, or conversion into N-substituted thioureas, followed by an intramolecular cyclodesulfurization reaction promoted by yellow HgO, furnished such spirocycles in excellent yields. Title compounds were tested in vitro against a panel of six human tumor cell lines, named A549 (non-small cell lung), HBL-100 (breast), HeLa (cervix), SW1573 (non-small cell lung), T-47D (breast) and WiDr (colon), and the results were compared with steroidal chemotherapeutic agents (abiraterone and galeterone); the A-ring of the steroidal backbone, the nature of the heterocycle and the N-substituents proved to be essential motifs for establishing structure-activity relationships concerning not only the potency but also the selectivity against tumor cell lines. Estrone derivatives, particularly those bearing a spiranic 2-aminooxazoline scaffold were found to be the most active compounds, with GI50 values ranging from the low micromolar to the submicromolar level (0.34-1.5 μM). Noteworthy, the lead compounds showed a remarkable increase in activity against the resistant cancer cell lines (T-47D and WiDr) compared to the anticancer reference drugs (up to 120-fold).
Collapse
Affiliation(s)
- Laura L Romero-Hernández
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, 72570 Puebla, Pue., Mexico
| | - Penélope Merino-Montiel
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, 72570 Puebla, Pue., Mexico.
| | - Socorro Meza-Reyes
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, 72570 Puebla, Pue., Mexico
| | - José Luis Vega-Baez
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, 72570 Puebla, Pue., Mexico
| | - Óscar López
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071 Seville, Spain
| | - José M Padrón
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Centro de Investigaciones Biomédicas de Canarias (CIBICAN), Universidad de La Laguna, c/ Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain
| | - Sara Montiel-Smith
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, 72570 Puebla, Pue., Mexico.
| |
Collapse
|