1
|
Hunt S, Thyagarajan A, Sahu RP. Dichloroacetate and Salinomycin as Therapeutic Agents in Cancer. Med Sci (Basel) 2025; 13:47. [PMID: 40407542 PMCID: PMC12101198 DOI: 10.3390/medsci13020047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 04/09/2025] [Accepted: 04/21/2025] [Indexed: 05/26/2025] Open
Abstract
Cancer is the second leading cause of mortality worldwide. Despite the available treatment options, a majority of cancer patients develop drug resistance, indicating the need for alternative approaches. Repurposed drugs, such as antiglycolytic and anti-microbial agents, have gained substantial attention as potential alternative strategies against different disease pathophysiologies, including lung cancer. To that end, multiple studies have suggested that the antiglycolytic dichloroacetate (DCA) and the antibiotic salinomycin (SAL) possess promising anticarcinogenic activity, attributed to their abilities to target the key metabolic enzymes, ion transport, and oncogenic signaling pathways involved in regulating cancer cell behavior, including cell survival and proliferation. We used the following searches and selection criteria. (1) Biosis and PubMed were used with the search terms dichloroacetate; salinomycin; dichloroacetate as an anticancer agent; salinomycin as an anticancer agent; dichloroacetate side effects; salinomycin side effects; salinomycin combination therapy; dichloroacetate combination therapy; and dichloroacetate or salinomycin in combination with other agents, including chemotherapy and tyrosine kinase inhibitors. (2) The exclusion criteria included not being related to the mechanisms of DCA and SAL or not focusing on their anticancer properties. (3) All the literature was sourced from peer-reviewed journals within a timeframe of 1989 to 2024. Importantly, experimental studies have demonstrated that both DCA and SAL exert promising anticarcinogenic properties, as well as having synergistic effects in combination with other therapeutic agents, against multiple cancer models. The goal of this review is to highlight the mechanistic workings and efficacy of DCA and SAL as monotherapies, and their combination with other therapeutic agents in various cancer models, with a major emphasis on non-small-cell lung cancer (NSCLC) treatment.
Collapse
Affiliation(s)
- Sunny Hunt
- Department of Chemistry and Biochemistry, Oberlin College, 173 W Lorain St, Oberlin, OH 44074, USA;
| | - Anita Thyagarajan
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Dayton, OH 45435, USA;
| | - Ravi P. Sahu
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Dayton, OH 45435, USA;
| |
Collapse
|
2
|
Zeltser N, Zhu C, Oh J, Li CH, Boutros PC. Sex Differences in Cancer Functional Genomics: Gene Dependency and Drug Sensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636540. [PMID: 39975298 PMCID: PMC11838570 DOI: 10.1101/2025.02.05.636540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Patient sex influences a wide range of cancer phenotypes, including prevalence, response to therapy and survival endpoints. Molecular sex differences have been identified at all levels of the central dogma. It is hypothesized that these molecular differences may drive the observed clinical sex differences. Yet despite a growing catalog of molecular sex differences in a range of cancer types, their specific functional consequences remain unclear. To directly assess how patient sex impacts cancer cell function, we evaluated 1,209 cell lines subjected to CRISPR knockout, RNAi knockdown or drug exposures. Despite limited statistical power, we identified pan- and per-cancer sex differences in gene essentiality in six sex-linked and fourteen autosomal genes, and in drug sensitivity for two compounds. These data fill a gap in our understanding of the link between sex-differential molecular effects and patient phenotypes. They call for much more careful and systematic consideration of sex-specific effects in mechanistic and functional studies.
Collapse
Affiliation(s)
- Nicole Zeltser
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Chenghao Zhu
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Jieun Oh
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Constance H. Li
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Paul C. Boutros
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Antoszczak M, Mielczarek-Puta M, Struga M, Huczynski A. Urea and Thiourea Derivatives of Salinomycin as Agents Targeting Malignant Colon Cancer Cells. Anticancer Agents Med Chem 2025; 25:330-338. [PMID: 39390831 DOI: 10.2174/0118715206322603241002064435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Since it was discovered that a natural polyether ionophore called salinomycin (SAL) selectively inhibits human cancer cells, the scientific world has been paying special attention to this compound. It has been studied for nearly 15 years. OBJECTIVE Thus, a very interesting research direction is the chemical modification of SAL structure, which could give more biologically active agents. METHODS We evaluated the anticancer activity of (thio)urea analogues class of C20-epi-aminosalinomycin (compound 3b). The studies covered the generation of reactive oxygen species (ROS), proapoptotic activity, cytotoxic activity, and lipid peroxidation in vitro. RESULTS Thioureas 5a-5d showed antiproliferative activity against selected human colon cancer cell lines greater than that of chemically unmodified SAL, with a 2~10-fold higher potency towards a metastatic variant of colon cancer cells (SW620). Mechanistically, SAL derivatives showed proapoptotic activity in primary colon cancer cells and induced the production of reactive oxygen species (ROS) in these cells. In SW620 cells, SAL derivatives increased lipid peroxidation with a weak effect on apoptosis and low ROS formation with cytotoxic effects followed by cytostatic ones, suggesting different modes of action of the compounds against primary and metastatic colon cancer cells. CONCLUSION The results of this study suggested that urea and thiourea derivatives of SAL provide promising leads for the rational development of new anticancer active agents.
Collapse
Affiliation(s)
- Michal Antoszczak
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, Poznań, 61-614, Poland
| | - Magdalena Mielczarek-Puta
- Chair and Department of Biochemistry, Medical University of Warsaw, Banacha 1, Warszawa, 02-097, Poland
| | - Marta Struga
- Chair and Department of Biochemistry, Medical University of Warsaw, Banacha 1, Warszawa, 02-097, Poland
| | - Adam Huczynski
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, Poznań, 61-614, Poland
| |
Collapse
|
4
|
Dhiman A, Rana D, Benival D, Garkhal K. Comprehensive insights into glioblastoma multiforme: drug delivery challenges and multimodal treatment strategies. Ther Deliv 2025; 16:87-115. [PMID: 39445563 PMCID: PMC11703381 DOI: 10.1080/20415990.2024.2415281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and malignant brain tumors, with a high prevalence in elderly population. Most chemotherapeutic agents fail to reach the tumor site due to various challenges. However, smart nanocarriers have demonstrated excellent drug-loading capabilities, enabling them to cross the blood brain tumor barrier for the GBM treatment. Surface modification of nanocarriers has significantly enhanced their potential for targeting therapeutics. Moreover, recent innovations in drug therapies, such as the incorporation of theranostic agents in nanocarriers and antibody-drug conjugates, have offered newer insights for both diagnosis and treatment. This review focuses on recent advances in new therapeutic interventions for GBM, with an emphasis on the nanotheranostics systems to maximize therapeutic and diagnostic outcomes.
Collapse
Affiliation(s)
- Ashish Dhiman
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Dhwani Rana
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Derajram Benival
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Kalpna Garkhal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| |
Collapse
|
5
|
Jiang R, Zhang X, Li N, Mao Y, Chen H, Deng Z, Wang W, Jiang ZX, Xu L, Yang Z. Effective Synthesis of C20-Epi-Isothiocyanato-Salinomycin and its Thiourea Derivatives as Potential Anticancer Agents. Chemistry 2024; 30:e202402483. [PMID: 39316423 DOI: 10.1002/chem.202402483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/30/2024] [Accepted: 09/24/2024] [Indexed: 09/25/2024]
Abstract
Salinomycin, a naturally occurring polyether ionophore antibiotic isolated from Streptomyces albus, has been demonstrated potent cytotoxic activity against a variety of cancer cell lines. In particular, it exhibits selective targeting of cancer stem cells. However, systemic toxicity, drug resistance and low bioavailability of the drug significantly limit its potential applications. In this study, the C20-epi-isothiocyanate of salinomycin was designed and synthesized, and then reacted with amines as a versatile synthon to assemble a series of salinomycin thiourea derivatives, which improved the druggability of salinomycin. The antiproliferative activities of the compounds were evaluated in vitro against A549, HepG2, HeLa, 4T1, and MCF-7 cancer cell lines using the CCK-8 assay. The pharmacological results showed that some salinomycin thiourea derivatives exhibited excellent inhibitory activity against at least one of the tested tumor cells and high selectivity. Further mechanistic studies showed that compound 9 f, containing a 3,5-difluorobenzyl moiety, could directly induce apoptosis, probably by increasing caspase-9 protein expression and cell cycle arrest in G1 phase in a concentration dependent manner.
Collapse
Affiliation(s)
- Rui Jiang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xin Zhang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Na Li
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yuyin Mao
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Huan Chen
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Zhouming Deng
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wentao Wang
- CAS Key Laboratory of Science and Technology on Applied Catalysis, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Zhong-Xing Jiang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, 430071, China
| | - Liying Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhigang Yang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
6
|
Allen-Taylor D, Boro G, Cabato P, Mai C, Nguyen K, Rijal G. Staphylococcus epidermidis biofilm in inflammatory breast cancer and its treatment strategies. Biofilm 2024; 8:100220. [PMID: 39318870 PMCID: PMC11420492 DOI: 10.1016/j.bioflm.2024.100220] [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: 08/06/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024] Open
Abstract
Bacterial biofilms represent a significant challenge in both clinical and industrial settings because of their robust nature and resistance to antimicrobials. Biofilms are formed by microorganisms that produce an exopolysaccharide matrix, protecting function and supporting for nutrients. Among the various bacterial species capable of forming biofilms, Staphylococcus epidermidis, a commensal organism found on human skin and mucous membranes, has emerged as a prominent opportunistic pathogen, when introduced into the body via medical devices, such as catheters, prosthetic joints, and heart valves. The formation of biofilms by S. epidermidis on these surfaces facilitates colonization and provides protection against host immune responses and antibiotic therapies, leading to persistent and difficult-to-treat infections. The possible involvement of biofilms for breast oncogenesis has recently created the curiosity. This paper therefore delves into S. epidermidis biofilm involvement in breast cancer. S. epidermidis biofilms can create a sustained inflammatory environment through their metabolites and can break DNA in breast tissue, promoting cellular proliferation, angiogenesis, and genetic instability. Preventing biofilm formation primarily involves preventing bacterial proliferation using prophylactic measures and sterilization of medical devices and equipment. In cancer treatment, common modalities include chemotherapy, surgery, immunotherapy, alkylating agents, and various anticancer drugs. Understanding the relationship between anticancer drugs and bacterial biofilms is crucial, especially for those undergoing cancer treatment who may be at increased risk of bacterial infections, for improving patient outcomes. By elucidating these interactions, strategies to prevent or disrupt biofilm formation, thereby reducing the incidence of infections associated with medical devices and implants, can be identified.
Collapse
Affiliation(s)
- D. Allen-Taylor
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| | - G. Boro
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| | - P.M. Cabato
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| | - C. Mai
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| | - K. Nguyen
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| | - G. Rijal
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| |
Collapse
|
7
|
Cao PHA, Dominic A, Lujan FE, Senthilkumar S, Bhattacharya PK, Frigo DE, Subramani E. Unlocking ferroptosis in prostate cancer - the road to novel therapies and imaging markers. Nat Rev Urol 2024; 21:615-637. [PMID: 38627553 PMCID: PMC12067944 DOI: 10.1038/s41585-024-00869-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 04/19/2024]
Abstract
Ferroptosis is a distinct form of regulated cell death that is predominantly driven by the build-up of intracellular iron and lipid peroxides. Ferroptosis suppression is widely accepted to contribute to the pathogenesis of several tumours including prostate cancer. Results from some studies reported that prostate cancer cells can be highly susceptible to ferroptosis inducers, providing potential for an interesting new avenue of therapeutic intervention for advanced prostate cancer. In this Perspective, we describe novel molecular underpinnings and metabolic drivers of ferroptosis, analyse the functions and mechanisms of ferroptosis in tumours, and highlight prostate cancer-specific susceptibilities to ferroptosis by connecting ferroptosis pathways to the distinctive metabolic reprogramming of prostate cancer cells. Leveraging these novel mechanistic insights could provide innovative therapeutic opportunities in which ferroptosis induction augments the efficacy of currently available prostate cancer treatment regimens, pending the elimination of major bottlenecks for the clinical translation of these treatment combinations, such as the development of clinical-grade inhibitors of the anti-ferroptotic enzymes as well as non-invasive biomarkers of ferroptosis. These biomarkers could be exploited for diagnostic imaging and treatment decision-making.
Collapse
Affiliation(s)
- Pham Hong Anh Cao
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Abishai Dominic
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fabiola Ester Lujan
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Sanjanaa Senthilkumar
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Mayo Clinic Alix School of Medicine, Rochester, MN, USA
| | - Pratip K Bhattacharya
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Center for Nuclear Receptors and Cell Signalling, University of Houston, Houston, TX, USA.
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.
| | - Elavarasan Subramani
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
8
|
Dhawefi N, Jedidi S, Sammari H, Ayari A, Jridi M, Sebai H. Diospyros kaki fruit aqueous extract individual/combined with famotidine mitigates peptic ulcer induced by alcohol in rats. Toxicol Res (Camb) 2024; 13:tfae155. [PMID: 39345794 PMCID: PMC11427753 DOI: 10.1093/toxres/tfae155] [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: 01/10/2024] [Revised: 06/11/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
The present study was performed to evaluate the therapeutic impact of Diospyros kaki fruit aqueous extract (DKFAE) on ethanol induced peptic ulcer. The phytochemical studies of DKFAE were investigated using colorometric analysis. Gastric ulcer was induced by one dose of ethanol (5 ml/Kg, b.w) on 24 h empty stomach. Then, the plant extract (200, 400 mg/kg) was orally administrated for 2 weeks. Famotidine (FAM: 40 mg/kg, b.w.): a reference drug was also tested. The effect of mixture dose between the fruit extract and FAM (DKFAE, 50 mg/kg PC, p.o. + FAM, 50 mg/kg PC, p.o.) was also evaluated. One hour after induction of ulcer blood samples were collected, stomach acidity and volume, as well as lesion counts were measured, then stomach and intestine of scarified rats were subjected to biochemical, macroscopic and microscopic studies. Results showed that DKFAE exhibited an important antioxidant potential. In vivo, the results showed that alcohol induced gastric damage, improving oxidative stress markers level such as MDA and H2O2, gastric and intestinal calcium and free iron. The intoxication by ethanol also produce an inflammation occurred by high level of the C-reactive protein (CRP) and alkaline phosphatase (ALP) activity in plasma. In contrast, DKFAE and the mixture dose significantly protect against macroscopic and histological injuries, the secretory profile disturbances, lipid peroxidation, antioxidant enzymes activities and non enzymatic antioxidant level decrease induced by ethanol administration. More impressively, the mixture dose exerted the more excellent effect than DKFAE and famotidine each alone showing is possible synergism.
Collapse
Affiliation(s)
- Nourhène Dhawefi
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja 9000 BP n° 382, Tunisia
- Laboratory of Sylvo-Pastoral Resources, University of Jendouba, Sylvo- Pastoral Institute of Tabarka, Tabarka 8110 Bp n° 345, Tunisia
| | - Saber Jedidi
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja 9000 BP n° 382, Tunisia
- Laboratory of Sylvo-Pastoral Resources, University of Jendouba, Sylvo- Pastoral Institute of Tabarka, Tabarka 8110 Bp n° 345, Tunisia
| | - Houcem Sammari
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja 9000 BP n° 382, Tunisia
- Laboratory of Sylvo-Pastoral Resources, University of Jendouba, Sylvo- Pastoral Institute of Tabarka, Tabarka 8110 Bp n° 345, Tunisia
| | - Ala Ayari
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja 9000 BP n° 382, Tunisia
| | - Mourad Jridi
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja 9000 BP n° 382, Tunisia
| | - Hichem Sebai
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja 9000 BP n° 382, Tunisia
| |
Collapse
|
9
|
Alhajamee M, Khalaj-Kondori M, Babaei E, Mahdavi M. A biochemical assessment of apoptosis-inducing impact of Salinomycin in combination with ciprofloxacin on human leukemia KG1-a stem-like cells in the presence and absence of insulin. Mol Biol Rep 2024; 51:807. [PMID: 39002036 DOI: 10.1007/s11033-024-09768-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/01/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Acute Myeloid Leukemia (AML) is a fast-developing invading cancer that impacts the blood and bone marrow, marked by the rapid proliferation of abnormal white blood cells. Chemotherapeutic agents, a primary treatment for AML, encounter clinical limitations such as poor solubility and low bioavailability. Previous studies have highlighted antibiotics as effective in inducing cancer cell death and potentially preventing metastasis. Besides, insulin is known to activate the PI3K/Akt pathway, often disrupted in cancers, leading to enhanced cell survival and resistance to apoptosis. In light of the above-mentioned points, we examined the anti-cancer impact of antibiotics Ciprofloxacin (CP) and Salinomycin (SAL) and their combination on KG1-a cells in the presence and absence of insulin. METHODS This was accomplished by exposing KG1-a cells to different doses of CP and SAL alone, in combination, and with or without insulin for 24-72 h. Cell viability was evaluated using the MTT assay. Besides, apoptotic effects were examined using Hoechst staining and Annexin-V/PI flow cytometry. The expression levels of Bax, p53, BIRC5, Akt, PTEN, and FOXO1 were analyzed through Real-Time PCR. RESULTS CP and SAL demonstrated cytotoxic and notable pro-apoptotic impact on KG1-a cells by upregulating Bax and p53 and downregulating BIRC5, leading to G0/G1 cell cycle arrest and prevention of the PI3K-Akt signaling pathway. Our findings demonstrated that combination of CP and SAL promote apoptosis in the KG1-a cell line by down-regulating BIRC5 and Akt, as well as up-regulating Bax, p53, PTEN, and FOXO1. Additionally, the findings strongly indicated that insulin effectively mitigates apoptosis by enhancing Akt expression and reducing FOXO1 and PTEN gene expression in the cells treated with CP and SAL. CONCLUSION Our findings showed that the combined treatment of CP and SAL exhibit a strong anti-cancer effect on leukemia KG1-a cells. Moreover, it was discovered that the PI3K-Akt signaling can be a promising target in leukemia treatment particularly in hyperinsulinemia condition.
Collapse
Affiliation(s)
- Maitham Alhajamee
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | - Esmaeil Babaei
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Majid Mahdavi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
10
|
Abroon S, Nouri M, Mahdavi M. Hesperidin/Salinomycin Combination; a Natural Product for Deactivation of the PI3K/Akt Signaling Pathway and Anti-Apoptotic Factors in KG1a Cells. J Fluoresc 2024:10.1007/s10895-024-03808-4. [PMID: 38916633 DOI: 10.1007/s10895-024-03808-4] [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: 03/27/2024] [Accepted: 06/06/2024] [Indexed: 06/26/2024]
Abstract
AML is a highly aggressive malignant clonal disease of hematopoietic origin. Hesperidin as a polyphenol glycoside, Activates the apoptotic pathway and salinomycin as a k + selective ionophore. We examined how hesperidin and salinomycin induce pro-apoptotic effects in KG1a cells. Cells were divided into four groups; 1) control cells (CRTL), 2) cells treated with hesperidin 85 μM, 3) cells treated with 2 μM salinomycin, 4) cells treated with combination of salinomycin and hesperidin. The MTT assay was implemented to determine the IC50 of hesperidin and salinomycin in KG1a cell lines. Propidium iodide staining and flow cytometry were used to analyze the distribution of the cell cycle. The level of ROS was evaluated by fluorescent microscopy and spectrophotometry. Additionally, Akt, XIAP, Bad, and FOXO1 gene expression was analyzed by real-time PCR. Hesperidin/Salinomycin decreased the viability of KG1a leukemic cells more than Hesperidin and Salinomycin separately. Changes in the shape of apoptotic cells and rise in ROS levels were detected after Hesperidin/Salinomycin treatment. Our findings showed that following Hesperidin/Salinomycin treatment, the expression of PI3K/AKT signaling pathway related genes (AKT, PTEN and FOXO1), were in line with the destruction of KG-1a cells. Furthermore, XIAP and BAD mRNA were regulated to trigger apoptosis in cancer cells. The study discovered that hesperidin and salinomycin, could effectively hinder the PI3K/Akt signaling pathway in leukemia cancer cells. Also, the combination of hesperidin and salinomycin has the potential to be a treatment option for acute myeloid leukemia.
Collapse
Affiliation(s)
- Sina Abroon
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
- Department of biochemistry and clinical laboratories, Faculty of medicine, Tabriz University of medical sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of biochemistry and clinical laboratories, Faculty of medicine, Tabriz University of medical sciences, Tabriz, Iran
| | - Majid Mahdavi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
11
|
Bano N, Parveen S, Saeed M, Siddiqui S, Abohassan M, Mir SS. Drug Repurposing of Selected Antibiotics: An Emerging Approach in Cancer Drug Discovery. ACS OMEGA 2024; 9:26762-26779. [PMID: 38947816 PMCID: PMC11209889 DOI: 10.1021/acsomega.4c00617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/19/2024] [Accepted: 05/29/2024] [Indexed: 07/02/2024]
Abstract
Drug repurposing is a method of investigating new therapeutic applications for previously approved medications. This repurposing approach to "old" medications is now highly efficient, simple to arrange, and cost-effective and poses little risk of failure in treating a variety of disorders, including cancer. Drug repurposing for cancer therapy is currently a key topic of study. It is a way of exploring recent therapeutic applications for already-existing drugs. Theoretically, the repurposing strategy has various advantages over the recognized challenges of creating new molecular entities, including being faster, safer, easier, and less expensive. In the real world, several medications have been repurposed, including aspirin, metformin, and chloroquine. However, doctors and scientists address numerous challenges when repurposing drugs, such as the fact that most drugs are not cost-effective and are resistant to bacteria. So the goal of this review is to gather information regarding repurposing pharmaceuticals to make them more cost-effective and harder for bacteria to resist. Cancer patients are more susceptible to bacterial infections. Due to their weak immune systems, antibiotics help protect them from a variety of infectious diseases. Although antibiotics are not immune boosters, they do benefit the defense system by killing bacteria and slowing the growth of cancer cells. Their use also increases the therapeutic efficacy and helps avoid recurrence. Of late, antibiotics have been repurposed as potent anticancer agents because of the evolutionary relationship between the prokaryotic genome and mitochondrial DNA of eukaryotes. Anticancer antibiotics that prevent cancer cells from growing by interfering with their DNA and blocking growth of promoters, which include anthracyclines, daunorubicin, epirubicin, mitoxantrone, doxorubicin, and idarubicin, are another type of FDA-approved antibiotics used to treat cancer. According to the endosymbiotic hypothesis, prokaryotes and eukaryotes are thought to have an evolutionary relationship. Hence, in this study, we are trying to explore antibiotics that are necessary for treating diseases, including cancer, helping people reduce deaths associated with various infections, and substantially extending people's life expectancy and quality of life.
Collapse
Affiliation(s)
- Nilofer Bano
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow 226026, India
- Department
of Bioengineering, Faculty of Engineering, Integral University, Kursi Road, Lucknow 226026, India
| | - Sana Parveen
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow 226026, India
- Department
of Biosciences, Faculty of Science, Integral
University, Kursi Road, Lucknow 226026, India
| | - Mohd Saeed
- Department
of Biology, College of Sciences, University
of Hail, P.O. Box 2240, Hail 55476, Saudi Arabia
| | - Samra Siddiqui
- Department
of Health Services Management, College of Public Health and Health
Informatics, University of Hail, Hail 55476, Saudi Arabia
| | - Mohammad Abohassan
- Department
of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
| | - Snober S. Mir
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow 226026, India
- Department
of Biosciences, Faculty of Science, Integral
University, Kursi Road, Lucknow 226026, India
| |
Collapse
|
12
|
Li L, Zhao L, Yang J, Zhou L. Multifaceted effects of LRP6 in cancer: exploring tumor development, immune modulation and targeted therapies. Med Oncol 2024; 41:180. [PMID: 38898247 DOI: 10.1007/s12032-024-02399-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 04/26/2024] [Indexed: 06/21/2024]
Abstract
Low-density lipoprotein receptor (LDLR)-related protein 6 (LRP6), a member of the LDLR superfamily of cell surface receptors, is most widely known as a crucial co-receptor in the activation of canonical Wnt/β-catenin signaling. This signaling pathway is implicated in multiple biological processes, such as lipoprotein metabolism, protease regulation, cell differentiation, and migration. LRP6 is frequently overexpressed in a variety of tumors, including liver cancer, colorectal cancer, and prostate cancer, and is generally considered an oncogene that promotes tumor proliferation, migration, and invasion. However, there are exceptions; some studies have reported that LRP6 inhibits lung metastasis of breast cancer through its ectodomain (LRP6N), and patients with low LRP6 expression tend to have a poor prognosis. Thus, the role of LRP6 in tumors remains controversial. Although limited studies have shown that LRP6 is associated with the expression and roles of a variety of immune cells in tumors, the interaction of LRP6 with the tumor microenvironment (TME) is not fully understood. Furthermore, it is crucial to acknowledge that LRP6 can engage with alternative pathways, including the mTORC1, CXCL12/CXCR4, and KRAS signaling pathways mentioned earlier, resulting in the regulation of biological functions independent of canonical Wnt/β-catenin signaling. Due to the potential of LRP6 as a molecular target for cancer therapy, various treatment modalities have been developed to directly or indirectly inhibit LRP6 function, demonstrating promising anti-cancer effects across multiple cancer types. This review will concentrate on exploring the expression, function, and potential therapeutic applications of LRP6 in different cancer types, along with its influence on the TME.
Collapse
Affiliation(s)
- Liangliang Li
- Department of Hematology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Li Zhao
- Laboratory of Clinical Molecular Cytogenetics and Immunology, The First Hospital of Lanzhou University, Lanzhou, Gansu, People's Republic of China
- Gansu Key Laboratory of Genetic Study of Hematopathy, Lanzhou, Gansu, People's Republic of China
| | - Jincai Yang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Lanxia Zhou
- Laboratory of Clinical Molecular Cytogenetics and Immunology, The First Hospital of Lanzhou University, Lanzhou, Gansu, People's Republic of China.
- Gansu Key Laboratory of Genetic Study of Hematopathy, Lanzhou, Gansu, People's Republic of China.
| |
Collapse
|
13
|
Rahman MA, Apu EH, Rakib-Uz-Zaman SM, Chakraborti S, Bhajan SK, Taleb SA, Shaikh MH, Jalouli M, Harrath AH, Kim B. Exploring Importance and Regulation of Autophagy in Cancer Stem Cells and Stem Cell-Based Therapies. Cells 2024; 13:958. [PMID: 38891090 PMCID: PMC11171866 DOI: 10.3390/cells13110958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Autophagy is a globally conserved cellular activity that plays a critical role in maintaining cellular homeostasis through the breakdown and recycling of cellular constituents. In recent years, there has been much emphasis given to its complex role in cancer stem cells (CSCs) and stem cell treatment. This study examines the molecular processes that support autophagy and how it is regulated in the context of CSCs and stem cell treatment. Although autophagy plays a dual role in the management of CSCs, affecting their removal as well as their maintenance, the intricate interaction between the several signaling channels that control cellular survival and death as part of the molecular mechanism of autophagy has not been well elucidated. Given that CSCs have a role in the development, progression, and resistance to treatment of tumors, it is imperative to comprehend their biological activities. CSCs are important for cancer biology because they also show a tissue regeneration model that helps with organoid regeneration. In other words, the manipulation of autophagy is a viable therapeutic approach in the treatment of cancer and stem cell therapy. Both synthetic and natural substances that target autophagy pathways have demonstrated promise in improving stem cell-based therapies and eliminating CSCs. Nevertheless, there are difficulties associated with the limitations of autophagy in CSC regulation, including resistance mechanisms and off-target effects. Thus, the regulation of autophagy offers a versatile strategy for focusing on CSCs and enhancing the results of stem cell therapy. Therefore, understanding the complex interactions between autophagy and CSC biology would be essential for creating therapeutic treatments that work in both regenerative medicine and cancer treatment.
Collapse
Affiliation(s)
- Md Ataur Rahman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Global Biotechnology and Biomedical Research Network (GBBRN), Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh
| | - Ehsanul Hoque Apu
- Department of Biomedical Sciences, College of Dental Medicine, Lincoln Memorial University, Knoxville, TN 37923, USA;
- DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN 37752, USA
- Division of Hematology and Oncology, Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - S. M Rakib-Uz-Zaman
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (S.M.R.-U.-Z.); (S.C.)
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, BRAC University, Dhaka 1212, Bangladesh
| | - Somdeepa Chakraborti
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (S.M.R.-U.-Z.); (S.C.)
| | - Sujay Kumar Bhajan
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science & Technology University, Gopalganj 8100, Bangladesh;
| | - Shakila Afroz Taleb
- Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06510, USA;
| | - Mushfiq H. Shaikh
- Department of Otolaryngology—Head and Neck Surgery, Western University, London, ON N6A 4V2, Canada;
| | - Maroua Jalouli
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia;
| | - Abdel Halim Harrath
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, 1-5 Hoegidong, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
14
|
Martyniuk V, Matskiv T, Yunko K, Khoma V, Gnatyshyna L, Faggio C, Stoliar O. Reductive stress and cytotoxicity in the swollen river mussel (Unio tumidus) exposed to microplastics and salinomycin. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 350:123724. [PMID: 38462197 DOI: 10.1016/j.envpol.2024.123724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/12/2024]
Abstract
Multistress effects lead to unpredicted consequences in aquatic ecotoxicology and are extremely concerning. The goal of this study was to trace how specific effects of the antibiotic salinomycin (Sal) and microplastics (MP) on the bivalve molluscs are manifested in the combined environmentally relevant exposures. Unio tumidus specimens were treated with Sal (0.6 μg L-1), MP (1 mg L-1, 2 μm size), and both at 18 °C (Mix) and 25 °C (MixT) for 14 days. The redox stress and apoptotic enzyme responses and the balance of Zn/Cu in the digestive gland were analyzed. The shared signs of stress included a decrease in NAD+/NADH and Zn/Cu ratios and lysosomal integrity and an increase in Zn-metallothioneins and cholinesterase levels. MP caused a decrease in the glutathione (GSH) concentration and redox state, total antioxidant capacity, and Zn levels. MP and Mix induced coordinated apoptotic/autophagy activities, increasing caspase-3 and cathepsin D (CtD) total and extralysosomal levels. Sal activated caspase-3 only and increased by five times Cu level in the tissue. Due to the discriminant analysis, the cumulative effect was evident in the combined exposure at 18 °C. However, under heating, the levels of NAD+, NADH, GSH, GSH/GSSG and metallothionein-related thiols were decreased, and coordination of the cytosolic and lysosomal death stimuli was distorted, confirming that heating and pollution could exert unexpected synergistic effects on aquatic life.
Collapse
Affiliation(s)
- Viktoria Martyniuk
- Department of Chemistry and Methods of Its Teaching, Ternopil Volodymyr Hnatiuk National Pedagogical University, Kryvonosa Str 2, Ternopil, 46027, Ukraine.
| | - Tetiana Matskiv
- Department of Chemistry and Methods of Its Teaching, Ternopil Volodymyr Hnatiuk National Pedagogical University, Kryvonosa Str 2, Ternopil, 46027, Ukraine; Department of General Chemistry, I. Horbachevsky Ternopil National Medical University, Maidan Voli, 1, Ternopil, 46001, Ukraine.
| | - Kateryna Yunko
- Department of Chemistry and Methods of Its Teaching, Ternopil Volodymyr Hnatiuk National Pedagogical University, Kryvonosa Str 2, Ternopil, 46027, Ukraine.
| | - Vira Khoma
- Department of Research of Materials, Substances and Products, Ternopil Scientific Research Forensic Center of the Ministry of Internal Affairs of Ukraine, St. Budny, 48, Ternopil, 46020, Ukraine.
| | - Lesya Gnatyshyna
- Department of General Chemistry, I. Horbachevsky Ternopil National Medical University, Maidan Voli, 1, Ternopil, 46001, Ukraine.
| | - Caterina Faggio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres, S. Agata, Messina, 31-98166, Italy; Department of Eco-sustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Naples, Italy.
| | - Oksana Stoliar
- Department of Chemistry and Methods of Its Teaching, Ternopil Volodymyr Hnatiuk National Pedagogical University, Kryvonosa Str 2, Ternopil, 46027, Ukraine; Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres, S. Agata, Messina, 31-98166, Italy.
| |
Collapse
|
15
|
Kim DR, Jeon CW, Kwak YS. Antifungal Properties of Streptomyces bacillaris S8 for Biological Control Applications. THE PLANT PATHOLOGY JOURNAL 2024; 40:322-328. [PMID: 38835303 PMCID: PMC11162865 DOI: 10.5423/ppj.nt.01.2024.0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 06/06/2024]
Abstract
Soybean (Glycine max), a crucial global crop, experiences yearly yield reduction due to diseases such as anthracnose (Colletotrichum truncatum) and root rot (Fusarium spp.). The use of fungicides, which have traditionally been employed to control these phytopathogens, is now facing challenges due to the emergence of fungicide-resistant strains. Streptomyces bacillaris S8 strain S8 is previously known to produce valinomycin t through a nonribosomal peptide synthetase (NRPS) pathway. The objective of this study was to evaluate the antifungal activity of S. bacillaris S8 against C. truncatum and Fusarium sp., assessing its efficacy against soybean pathogens. The results indicate that strain S8 effectively controlled both above-ground and underground soybean diseases, using the NRPS and NRPS-related compound, suggesting its potential as a biological control in plant-microbe interactions. These findings underscore the pivotal role of the stain S8 in fostering healthy soybean microbial communities and emphasize the significance of microbiota structure studies in unveiling potent biocontrol agents.
Collapse
Affiliation(s)
- Da-Ran Kim
- Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea
| | - Chang-Wook Jeon
- Division of Applied Life Science (BK21Plus), Gyeongsang National University, Jinju 52828, Korea
| | - Youn-Sig Kwak
- Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea
- Division of Applied Life Science (BK21Plus), Gyeongsang National University, Jinju 52828, Korea
| |
Collapse
|
16
|
Kanchan S, Marwaha D, Tomar B, Agrawal S, Mishra S, Kapoor R, Sushma, Jha G, Sharma D, Bhatta RS, Mishra PR, Rath SK. Nanocarrier - Mediated Salinomycin Delivery Induces Apoptosis and Alters EMT Phenomenon in Prostate Adenocarcinoma. AAPS PharmSciTech 2024; 25:104. [PMID: 38724836 DOI: 10.1208/s12249-024-02817-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/22/2024] [Indexed: 09/05/2024] Open
Abstract
Salinomycin (Sal) has been recently discovered as a novel chemotherapeutic agent against various cancers including prostate cancer which is one of the most commonly diagnosed cancers affecting male populations worldwide. Herein we designed salinomycin nanocarrier (Sal-NPs) to extend its systemic circulation and to increase its anticancer potential. Prepared nanoform showed high encapsulation and sustained release profile for salinomycin. The present study elucidated the cytotoxicity and mechanism of apoptotic cell death of Sal-NPs against prostate cancer both in vitro and in vivo. At all measured concentrations, Sal-NPs showed more significant cytotoxicity to DU145 and PC3 cells than Sal alone. This effect was mediated by apoptosis, as confirmed by ROS generation, loss of MMP and cell cycle arrest at the G1 phase in both cells. Sal-NPs efficiently inhibited migration of PC3 and DU145 cells via effectively downregulating the epithelial mesenchymal transition. Also, the results confirmed that Sal-NPs can effectively inhibit the induction of Prostate adenocarcinoma in male Wistar rats. Sal-NPs treatment exhibited a decrease in tumour sizes, a reduction in prostate weight, and an increase in body weight, which suggests that Sal-NPs is more effective than salinomycin alone. Our results suggest that the molecular mechanism underlying the Sal-NPs anticancer effect may lead to the development of a potential therapeutic strategy for treating prostate adenocarcinoma.
Collapse
Affiliation(s)
- Sonam Kanchan
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Bhawna Tomar
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sristi Agrawal
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sakshi Mishra
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Radhika Kapoor
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Sushma
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Gaurav Jha
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Divyansh Sharma
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Rabi Sankar Bhatta
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Srikanta Kumar Rath
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
17
|
Yu S, Tong L, Shen J, Li C, Hu Y, Feng K, Shao J. Recent research progress based on ferroptosis-related signaling pathways and the tumor microenvironment on it effects. Eur J Med Chem 2024; 269:116290. [PMID: 38518522 DOI: 10.1016/j.ejmech.2024.116290] [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: 12/18/2023] [Revised: 02/19/2024] [Accepted: 02/25/2024] [Indexed: 03/24/2024]
Abstract
The existing therapies for cancer are not remote satisfactory due to drug-resistance in tumors that are malignant. There is a pressing necessity to take a step forward to develop innovative therapies that can complement current ones. Multiple investigations have demonstrated that ferroptosis therapy, a non-apoptotic modality of programmed cell death, has tremendous potential in face of multiple crucial events, such as drug resistance and toxicity in aggressive malignancies. Recently, ferroptosis at the crosswalk of chemotherapy, materials science, immunotherapy, tumor microenvironment, and bionanotechnology has been presented to elucidate its therapeutic feasibility. Given the burgeoning progression of ferroptosis-based nanomedicine, the newest advancements in this field at the confluence of ferroptosis-inducers, nanotherapeutics, along with tumor microenvironment are given an overview. Here, the signaling pathways of ferroptosis-related were first talked about briefly. The emphasis discussion was placed on the pharmacological mechanisms and the nanodrugs design of ferroptosis inducing agents based on multiple distinct metabolism pathways. Additionally, a comprehensive overview of the action mechanisms by which the tumor microenvironment influences ferroptosis was elaborately descripted. Finally, some limitations of current researches and future research directions were also deliberately discussed to provide details about therapeutic avenues for ferroptosis-related diseases along with the design of anti-drugs.
Collapse
Affiliation(s)
- Shijing Yu
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Lingwu Tong
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Jiangwen Shen
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Chenglei Li
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Yongshan Hu
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Keke Feng
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Jingwei Shao
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China.
| |
Collapse
|
18
|
Woo WS, Shim SH, Kang G, Kim KH, Son HJ, Sohn MY, Lee S, Kim J, Seo JS, Kwon MG, Kim DH, Park CI. Assessment of Salinomycin's Potential to Treat Microcotyle sebastis in Korean Rockfish ( Sebastes schlegelii). Animals (Basel) 2023; 13:3233. [PMID: 37893959 PMCID: PMC10603687 DOI: 10.3390/ani13203233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Aquaculture, a crucial sector of the global food industry, faces a myriad of issues due to parasitic invasions. One such parasite, Microcotyle sebastis, which afflicts Korean rockfish in South Korea, has a significant economic impact. The impending danger of resistance to traditional anthelmintics necessitates the exploration of new antiparasitic candidates. Although the efficacy of salinomycin against aquatic parasites such as ciliates and sporozoans is known, its influence on monogeneans has yet to be studied. Therefore, this study investigated the efficacy and safety of salinomycin for the treatment of M. sebastis infections, presenting the first exploration of salinomycin's therapeutic potential against monogeneans. In vitro examinations revealed a minimum effective concentration of salinomycin of 5 mg/kg, which led to necrosis of the haptor upon dislodging from the gill filaments. The one-time oral administration of the drug at concentrations of 5 mg/kg and 10 mg/kg showed a significant dose-dependent reduction in parasite counts, with no apparent behavioral side effects in Korean rockfish. Biochemical analyses monitored the liver, heart, and kidney enzymes, specifically aspartate transaminase (AST), alanine transaminase (ALT), blood urea nitrogen (BUN), and creatine kinase-myocardial band (CK-MB). At both 20 °C and 13 °C, no significant differences were observed in the levels of AST and ALT. However, at 20 °C, alterations in BUN levels were evident on Day 14, a deviation not observed at 13 °C. The CK-MB analysis revealed elevated enzyme levels at both temperatures when compared to the control group, reflecting the similar changes observed in terrestrial animals administered salinomycin. The biochemical data suggest that the oral administration of salinomycin is potentially more favorable at 13 °C than at 20 °C. Although our findings warrant further comprehensive studies, including on the long-term and potential effects on nontarget species and water quality, they also suggest that salinomycin could be considered as an alternative or adjunctive treatment if resistance to the currently used praziquantel against M. sebastis is confirmed.
Collapse
Affiliation(s)
- Won-Sik Woo
- Department of Marine Biology & Aquaculture, Institute of Marine Industry, College of Marine Science, Gyeongsang National University, 2 Tongyeonghaean-ro, Tongyeong 53064, Republic of Korea
| | - Sang Hee Shim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Gyoungsik Kang
- Department of Marine Biology & Aquaculture, Institute of Marine Industry, College of Marine Science, Gyeongsang National University, 2 Tongyeonghaean-ro, Tongyeong 53064, Republic of Korea
| | - Kyung-Ho Kim
- Department of Marine Biology & Aquaculture, Institute of Marine Industry, College of Marine Science, Gyeongsang National University, 2 Tongyeonghaean-ro, Tongyeong 53064, Republic of Korea
| | - Ha-Jeong Son
- Department of Marine Biology & Aquaculture, Institute of Marine Industry, College of Marine Science, Gyeongsang National University, 2 Tongyeonghaean-ro, Tongyeong 53064, Republic of Korea
| | - Min-Young Sohn
- Department of Marine Biology & Aquaculture, Institute of Marine Industry, College of Marine Science, Gyeongsang National University, 2 Tongyeonghaean-ro, Tongyeong 53064, Republic of Korea
| | - Seungjin Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaekyeong Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jung-Soo Seo
- Aquatic Disease Control Division, National Fishery Products Quality Management Service, 216 Gijanghaean-ro, Busan 46083, Republic of Korea
| | - Mun-Gyeong Kwon
- Aquatic Disease Control Division, National Fishery Products Quality Management Service, 216 Gijanghaean-ro, Busan 46083, Republic of Korea
| | - Do-Hyung Kim
- Department of Aquatic Life Medicine, College of Fisheries Sciences, Pukyong National University, Busan 48513, Republic of Korea
| | - Chan-Il Park
- Department of Marine Biology & Aquaculture, Institute of Marine Industry, College of Marine Science, Gyeongsang National University, 2 Tongyeonghaean-ro, Tongyeong 53064, Republic of Korea
| |
Collapse
|
19
|
Abstract
Covering: from 2000 up to the very early part of 2023S-Adenosyl-L-methionine (SAM) is a naturally occurring trialkyl sulfonium molecule that is typically associated with biological methyltransfer reactions. However, SAM is also known to donate methylene, aminocarboxypropyl, adenosyl and amino moieties during natural product biosynthetic reactions. The reaction scope is further expanded as SAM itself can be modified prior to the group transfer such that a SAM-derived carboxymethyl or aminopropyl moiety can also be transferred. Moreover, the sulfonium cation in SAM has itself been found to be critical for several other enzymatic transformations. Thus, while many SAM-dependent enzymes are characterized by a methyltransferase fold, not all of them are necessarily methyltransferases. Furthermore, other SAM-dependent enzymes do not possess such a structural feature suggesting diversification along different evolutionary lineages. Despite the biological versatility of SAM, it nevertheless parallels the chemistry of sulfonium compounds used in organic synthesis. The question thus becomes how enzymes catalyze distinct transformations via subtle differences in their active sites. This review summarizes recent advances in the discovery of novel SAM utilizing enzymes that rely on Lewis acid/base chemistry as opposed to radical mechanisms of catalysis. The examples are categorized based on the presence of a methyltransferase fold and the role played by SAM within the context of known sulfonium chemistry.
Collapse
Affiliation(s)
- Yu-Hsuan Lee
- Department of Chemistry, University of Texas at Austin, Austin, TX 78712, USA.
| | - Daan Ren
- Department of Chemistry, University of Texas at Austin, Austin, TX 78712, USA.
| | - Byungsun Jeon
- Department of Chemistry, University of Texas at Austin, Austin, TX 78712, USA.
| | - Hung-Wen Liu
- Department of Chemistry, University of Texas at Austin, Austin, TX 78712, USA.
- Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
20
|
Petkov N, Pantcheva I, Ivanova A, Stoyanova R, Kukeva R, Alexandrova R, Abudalleh A, Dorkov P. Novel Cerium(IV) Coordination Compounds of Monensin and Salinomycin. Molecules 2023; 28:4676. [PMID: 37375231 DOI: 10.3390/molecules28124676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
The largely uncharted complexation chemistry of the veterinary polyether ionophores, monensic and salinomycinic acids (HL) with metal ions of type M4+ and the known antiproliferative potential of antibiotics has provoked our interest in exploring the coordination processes between MonH/SalH and ions of Ce4+. (1) Methods: Novel monensinate and salinomycinate cerium(IV)-based complexes were synthesized and structurally characterized by elemental analysis, a plethora of physicochemical methods, density functional theory, molecular dynamics, and biological assays. (2) Results: The formation of coordination species of a general composition [CeL2(OH)2] and [CeL(NO3)2(OH)], depending on reaction conditions, was proven both experimentally and theoretically. The metal(IV) complexes [CeL(NO3)2(OH)] possess promising cytotoxic activity against the human tumor uterine cervix (HeLa) cell line, being highly selective (non-tumor embryo Lep-3 vs. HeLa) compared to cisplatin, oxaliplatin, and epirubicin.
Collapse
Affiliation(s)
- Nikolay Petkov
- Faculty of Chemistry and Pharmacy, Sofia University St. Kliment Ohridski, 1164 Sofia, Bulgaria
| | - Ivayla Pantcheva
- Faculty of Chemistry and Pharmacy, Sofia University St. Kliment Ohridski, 1164 Sofia, Bulgaria
| | - Anela Ivanova
- Faculty of Chemistry and Pharmacy, Sofia University St. Kliment Ohridski, 1164 Sofia, Bulgaria
| | - Radostina Stoyanova
- Institute of General and Inorganic Chemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Rositsa Kukeva
- Institute of General and Inorganic Chemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Radostina Alexandrova
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Abedullkader Abudalleh
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Petar Dorkov
- Research and Development Department, Biovet Ltd., 4550 Peshtera, Bulgaria
| |
Collapse
|
21
|
Li L, Zeng P, Yu L, Yang J, Man J, Zhou L, Zhao L. Salinomycin sodium exerts anti diffuse large B-cell lymphoma activity through inhibition of LRP6-mediated Wnt/β-catenin and mTORC1 signaling. Leuk Lymphoma 2023:1-10. [PMID: 37092573 DOI: 10.1080/10428194.2023.2202291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Low-density lipoprotein receptor-related protein-6 (LRP6) is overexpressed in various cancers. The small molecule salinomycin sodium inhibits LRP6. We observed a higher proportion of subjects with non-germinal center B (non-GCB) subtypes having high LRP6 expression than those with GCB subtypes by immunohistochemistry. The PCR and Western blot assays demonstrated increased LRP6 expression in non-GCB subtype cells. In addition, CCK-8 assays and transwell cell migration assays revealed that salinomycin sodium exhibited dose- and time-dependent inhibition of proliferation and migration in non-GCB subtype cells. Furthermore, Western blot assays showed that salinomycin sodium decreased the expression of Bcl2, while increasing the expression of Bax. Additionally, salinomycin sodium suppressed LRP6 expression, blocked LRP6 phosphorylation, and inhibited the Wnt/β-catenin and mTORC1 signaling pathways. Our results suggest that LRP6 is highly expressed in non-GCB subtype. Furthermore, salinomycin sodium inhibited LRP6 expression and the Wnt/β-catenin and mTORC1 signaling in non-GCB subtype cells, and displayed potent anticancer activity.
Collapse
Affiliation(s)
- Liangliang Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, Gansu, P.R. China
| | - Pengyun Zeng
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, Gansu, P.R. China
| | - Lili Yu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
- Department of Medical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, P.R. China
| | - Jincai Yang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
| | - Jiancheng Man
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
| | - Lanxia Zhou
- Central Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, P.R. China
- Gansu Key Laboratory of Genetic Study of Hematopathy, Lanzhou, Gansu, P.R. China
| | - Li Zhao
- Central Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, P.R. China
- Gansu Key Laboratory of Genetic Study of Hematopathy, Lanzhou, Gansu, P.R. China
| |
Collapse
|
22
|
Anees M, Mehrotra N, Tiwari S, Kumar D, Kharbanda S, Singh H. Polylactic acid based biodegradable hybrid block copolymeric nanoparticle mediated co-delivery of salinomycin and doxorubicin for cancer therapy. Int J Pharm 2023; 635:122779. [PMID: 36842520 DOI: 10.1016/j.ijpharm.2023.122779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 02/05/2023] [Accepted: 02/21/2023] [Indexed: 02/28/2023]
Abstract
Existence of cancer stem cells (CSCs) are primarily responsible for chemoresistance, cancer reoccurrence and treatment failure in cancer patients. Eliminating CSCs along with bulk tumor is a necessity to achieve complete cancer inhibition. Salinomycin (SAL) has potential to specifically target and kill CSCs through blocking their multiple pathways simultaneously. SAL has also been reported to improve anti-cancer efficacy of numerous chemo-based drugs when used in combination therapy. However, clinical use of SAL is restricted due to its high off targeted toxicity. Herein, we have developed a PLA based hybrid block copolymer for concomitant delivery of SAL and doxorubicin (DOX) with an aim to reduce their adverse side effects and enhance the therapeutic efficacy of the treatment. Designed PLA based nanoplatform showed high encapsulation and sustained release profile for both the drugs. Cytotoxicity evaluation on cancer cell lines confirmed the synergistic effect of SAL:DOX co-loaded NPs. Additionally, prepared SAL NPs were also found to be highly effective against chemo-resistant cancer cells and CSCs derived from cancer patient. Most importantly, encapsulation of SAL in PLA NPs improved its pharmacokinetics and biodistribution profile. Consequently, undesired toxicity with SAL NPs was significantly reduced which in-turn increased the dose tolerability in mice as compared to free SAL. Treatment of EAC tumor bearing mice with SAL:DOX co-loaded NPs resulted in excellent tumor regression and complete inhibition of cancer reoccurrence. These results conclude that concomitant delivery of SAL and DOX using PLA based block copolymeric nano-carrier have a strong potential for cancer therapy.
Collapse
Affiliation(s)
- Mohd Anees
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Neha Mehrotra
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Sachchidanand Tiwari
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Dinesh Kumar
- National Institute of Health and Family Welfare (NIHFW), New Delhi 110067, India
| | | | - Harpal Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India; All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
23
|
Martyniuk V, Khoma V, Matskiv T, Yunko K, Gnatyshyna L, Stoliar O, Faggio C. Combined effect of microplastic, salinomycin and heating on Unio tumidus. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 98:104068. [PMID: 36680920 DOI: 10.1016/j.etap.2023.104068] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/06/2023] [Accepted: 01/18/2023] [Indexed: 06/17/2023]
Abstract
Microplastic (MP) and heating (T) suspected to modulate biological effects of aquatic contaminants. Salinomycin (Sal) is veterinary antibiotic and anticancer agent. The goal of this study was to examine the multistress effect of MP, Sal and T on the bioindicator bivalve mollusc. The Unio tumidus were treated with MP (1 mg L-1), Sal (0.6 µg L-1), their combination under 18° C (Mix) and 25° C (MixT) for 14 days. The digestive glands were analyzed. MP and Sal did not cause changes of Mn- and Cu,Zn-SOD, lipid peroxidation and Cyp-450-depended EROD levels, whereas catalase, GST and protein carbonyls (Sal-group) increased compared to control. In the Mix-group, enzymes, particularly EROD and GST (by 34% and 115% respectively) were up-regulated. However, in the MixT-group, they were corresponding to control or lesser (EROD, catalase). Our findings emphasize the need to take into account multistress interactions in the MP environmental risk assessment.
Collapse
Affiliation(s)
- Viktoria Martyniuk
- Ternopil Volodymyr Hnatiuk National Pedagogical University, M. Kryvonosa Str. 2, 46027 Ternopil, Ukraine.
| | - Vira Khoma
- Ternopil Volodymyr Hnatiuk National Pedagogical University, M. Kryvonosa Str. 2, 46027 Ternopil, Ukraine.
| | - Tetiana Matskiv
- Ternopil Volodymyr Hnatiuk National Pedagogical University, M. Kryvonosa Str. 2, 46027 Ternopil, Ukraine; I. Ya. Horbachevsky Ternopil National Medical University, Maidan Voli 1, 46001 Ternopil, Ukraine.
| | - Kateryna Yunko
- Ternopil Volodymyr Hnatiuk National Pedagogical University, M. Kryvonosa Str. 2, 46027 Ternopil, Ukraine.
| | - Lesya Gnatyshyna
- Ternopil Volodymyr Hnatiuk National Pedagogical University, M. Kryvonosa Str. 2, 46027 Ternopil, Ukraine; I. Ya. Horbachevsky Ternopil National Medical University, Maidan Voli 1, 46001 Ternopil, Ukraine.
| | - Oksana Stoliar
- Ternopil Volodymyr Hnatiuk National Pedagogical University, M. Kryvonosa Str. 2, 46027 Ternopil, Ukraine.
| | - Caterina Faggio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres, 31, 98166 S Agata -Messina, Italy.
| |
Collapse
|
24
|
Koirala N, Butnariu M, Panthi M, Gurung R, Adhikari S, Subba RK, Acharya Z, Popović-Djordjević J. Antibiotics in the management of tuberculosis and cancer. ANTIBIOTICS - THERAPEUTIC SPECTRUM AND LIMITATIONS 2023:251-294. [DOI: 10.1016/b978-0-323-95388-7.00014-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
25
|
Potential Nanotechnology-Based Therapeutics to Prevent Cancer Progression through TME Cell-Driven Populations. Pharmaceutics 2022; 15:pharmaceutics15010112. [PMID: 36678741 PMCID: PMC9864587 DOI: 10.3390/pharmaceutics15010112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with a high risk of metastasis and therapeutic resistance. These issues are closely linked to the tumour microenvironment (TME) surrounding the tumour tissue. The association between residing TME components with tumour progression, survival, and metastasis has been well elucidated. Focusing on cancer cells alone is no longer considered a viable approach to therapy; thus, there is a high demand for TME targeting. The benefit of using nanoparticles is their preferential tumour accumulation and their ability to target TME components. Several nano-based platforms have been investigated to mitigate microenvironment-induced angiogenesis, therapeutic resistance, and tumour progression. These have been achieved by targeting mesenchymal originating cells (e.g., cancer-associated fibroblasts, adipocytes, and stem cells), haematological cells (e.g., tumour-associated macrophages, dendritic cells, and myeloid-derived suppressor cells), and the extracellular matrix within the TME that displays functional and architectural support. This review highlights the importance of nanotechnology-based therapeutics as a promising approach to target the TME and improve treatment outcomes for TNBC patients, which can lead to enhanced survival and quality of life. The role of different nanotherapeutics has been explored in the established TME cell-driven populations.
Collapse
|
26
|
Yin L, Liu P, Jin Y, Ning Z, Yang Y, Gao H. Ferroptosis-related small-molecule compounds in cancer therapy: Strategies and applications. Eur J Med Chem 2022; 244:114861. [DOI: 10.1016/j.ejmech.2022.114861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/07/2022] [Accepted: 10/17/2022] [Indexed: 01/17/2023]
|
27
|
Cheng X, Zheng H, Wang C, Wang X, Fei C, Zhou W, Zhang K. Effects of salinomycin and ethanamizuril on the three microbial communities in vivo and in vitro. Front Microbiol 2022; 13:941259. [PMID: 36033856 PMCID: PMC9413843 DOI: 10.3389/fmicb.2022.941259] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/01/2022] [Indexed: 11/24/2022] Open
Abstract
The fate of a drug is not only the process of drug metabolism in vivo and in vitro but also the homeostasis of drug-exposed microbial communities may be disturbed. Anticoccidial drugs are widely used to combat the detrimental effects of protozoan parasites in the poultry industry. Salinomycin and ethanamizuril belong to two different classes of anticoccidial drugs. The effect of salinomycin and ethanamizuril on the microbiota of cecal content, manure compost, and soil remains unknown. Our results showed that although both salinomycin and ethanamizuril treatments suppressed some opportunistic pathogens, they failed to repair the great changes in chicken cecal microbial compositions caused by coccidia infection. Subsequently, the metabolite5 profiling of cecal content by LC-MS/MS analyses confirmed the great impact of coccidia infection on chicken cecum and showed that histidine metabolism may be the main action pathway of salinomycin, and aminoacyl tRNA biosynthesis may be the major regulatory mechanism of ethanamizuril. The microbial community of manure compost showed a mild response to ethanamizuril treatment, but ethanamizuril in soil could promote Actinobacteria reproduction, which may inhibit other taxonomic bacteria. When the soil and manure were exposed to salinomycin, the Proteobacteria abundance of microbial communities showed a significant increase, which suggested that salinomycin may improve the ability of the microbiota to utilize carbon sources. This hypothesis was confirmed by a BIOLOG ECO microplate analysis. In the animal model of coccidia infection, the treatment of salinomycin and ethanamizuril may reconstruct a new equilibrium of the intestinal microbiota. In an in vitro environment, the effect of ethanamizuril on composting and soil microbiota seems to be slight. However, salinomycin has a great impact on the microbial communities of manure composting and soil. In particular, the promoting effect of salinomycin on Proteobacteria phylum should be further concerned. In general, salinomycin and ethanamizuril have diverse effects on various microbial communities.
Collapse
|
28
|
Properties of Leukemic Stem Cells in Regulating Drug Resistance in Acute and Chronic Myeloid Leukemias. Biomedicines 2022; 10:biomedicines10081841. [PMID: 36009388 PMCID: PMC9405586 DOI: 10.3390/biomedicines10081841] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Notoriously known for their capacity to reconstitute hematological malignancies in vivo, leukemic stem cells (LSCs) represent key drivers of therapeutic resistance and disease relapse, posing as a major medical dilemma. Despite having low abundance in the bulk leukemic population, LSCs have developed unique molecular dependencies and intricate signaling networks to enable self-renewal, quiescence, and drug resistance. To illustrate the multi-dimensional landscape of LSC-mediated leukemogenesis, in this review, we present phenotypical characteristics of LSCs, address the LSC-associated leukemic stromal microenvironment, highlight molecular aberrations that occur in the transcriptome, epigenome, proteome, and metabolome of LSCs, and showcase promising novel therapeutic strategies that potentially target the molecular vulnerabilities of LSCs.
Collapse
|
29
|
Dey P, Ray Chaudhuri S. Cancer-Associated Microbiota: From Mechanisms of Disease Causation to Microbiota-Centric Anti-Cancer Approaches. BIOLOGY 2022; 11:757. [PMID: 35625485 PMCID: PMC9138768 DOI: 10.3390/biology11050757] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/08/2022] [Accepted: 05/12/2022] [Indexed: 02/07/2023]
Abstract
Helicobacter pylori infection is the only well-established bacterial cause of cancer. However, due to the integral role of tissue-resident commensals in maintaining tissue-specific immunometabolic homeostasis, accumulated evidence suggests that an imbalance of tissue-resident microbiota that are otherwise considered as commensals, can also promote various types of cancers. Therefore, the present review discusses compelling evidence linking tissue-resident microbiota (especially gut bacteria) with cancer initiation and progression. Experimental evidence supporting the cancer-causing role of gut commensal through the modulation of host-specific processes (e.g., bile acid metabolism, hormonal effects) or by direct DNA damage and toxicity has been discussed. The opportunistic role of commensal through pathoadaptive mutation and overcoming colonization resistance is discussed, and how chronic inflammation triggered by microbiota could be an intermediate in cancer-causing infections has been discussed. Finally, we discuss microbiota-centric strategies, including fecal microbiota transplantation, proven to be beneficial in preventing and treating cancers. Collectively, this review provides a comprehensive understanding of the role of tissue-resident microbiota, their cancer-promoting potentials, and how beneficial bacteria can be used against cancers.
Collapse
Affiliation(s)
- Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Saumya Ray Chaudhuri
- Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology, Chandigarh 160036, India;
| |
Collapse
|
30
|
Dudev T, Cheshmedzhieva D, Dorkov P, Pantcheva I. A DFT/PCM Study on the Affinity of Salinomycin to Bind Monovalent Metal Cations. Molecules 2022; 27:532. [PMID: 35056843 PMCID: PMC8779476 DOI: 10.3390/molecules27020532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/08/2022] [Accepted: 01/11/2022] [Indexed: 11/29/2022] Open
Abstract
The affinity of the polyether ionophore salinomycin to bind IA/IB metal ions was accessed using the Gibbs free energy of the competition reaction between SalNa (taken as a reference) and its rival ions: [M+-solution] + [SalNa] → [SalM] + [Na+-solution] (M = Li, K, Rb, Cs, Cu, Ag, Au). The DFT/PCM computations revealed that the ionic radius, charge density and accepting ability of the competing metal cations, as well as the dielectric properties of the solvent, have an influence upon the selectivity of salinomycin. The optimized structures of the monovalent metal complexes demonstrate the flexibility of the ionophore, allowing the coordination of one or two water ligands in SalM-W1 and SalM-W2, respectively. The metal cations are responsible for the inner coordination sphere geometry, with coordination numbers spread between 2 (Au+), 4 (Li+ and Cu+), 5/6 (Na+, K+, Ag+), 6/7 (Rb+) and 7/8 (Cs+). The metals' affinity to salinomycin in low-polarity media follows the order of Li+ > Cu+ > Na+ > K+ > Au+ > Ag+ > Rb+ > Cs+, whereas some derangement takes place in high-dielectric environment: Li+ ≥ Na+ > K+ > Cu+ > Au+ > Ag+ > Rb+ > Cs+.
Collapse
Affiliation(s)
- Todor Dudev
- Laboratory of Computational Chemistry and Spectroscopy, Faculty of Chemistry and Pharmacy, “St. Kl. Ohridski” University of Sofia, 1164 Sofia, Bulgaria;
| | - Diana Cheshmedzhieva
- Laboratory of Computational Chemistry and Spectroscopy, Faculty of Chemistry and Pharmacy, “St. Kl. Ohridski” University of Sofia, 1164 Sofia, Bulgaria;
| | - Peter Dorkov
- Research & Development Department, Biovet Ltd., 4550 Peshtera, Bulgaria;
| | - Ivayla Pantcheva
- Laboratory of Biocoordination and Bioanalytical Chemistry, Faculty of Chemistry and Pharmacy, “St. Kl. Ohridski” University of Sofia, 1164 Sofia, Bulgaria
| |
Collapse
|
31
|
Uncovering drug repurposing candidates for head and neck cancers: insights from systematic pharmacogenomics data analysis. Sci Rep 2021; 11:23933. [PMID: 34907286 PMCID: PMC8671460 DOI: 10.1038/s41598-021-03418-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 12/02/2021] [Indexed: 12/24/2022] Open
Abstract
Effective treatment options for head and neck squamous cell carcinoma (HNSCC) are currently lacking. We exploited the drug response and genomic data of the 28 HNSCC cell lines, screened with 4,518 compounds, from the PRISM repurposing dataset to uncover repurposing drug candidates for HNSCC. A total of 886 active compounds, comprising of 418 targeted cancer, 404 non-oncology, and 64 chemotherapy compounds were identified for HNSCC. Top classes of mechanism of action amongst targeted cancer compounds included PI3K/AKT/MTOR, EGFR, and HDAC inhibitors. We have shortlisted 36 compounds with enriched killing activities for repurposing in HNSCC. The integrative analysis confirmed that the average expression of EGFR ligands (AREG, EREG, HBEGF, TGFA, and EPGN) is associated with osimertinib sensitivity. Novel putative biomarkers of response including those involved in immune signalling and cell cycle were found to be associated with sensitivity and resistance to MEK inhibitors respectively. We have also developed an RShiny webpage facilitating interactive visualization to fuel further hypothesis generation for drug repurposing in HNSCC. Our study provides a rich reference database of HNSCC drug sensitivity profiles, affording an opportunity to explore potential biomarkers of response in prioritized drug candidates. Our approach could also reveal insights for drug repurposing in other cancers.
Collapse
|
32
|
Wang Q, Yen YT, Xie C, Liu F, Liu Q, Wei J, Yu L, Wang L, Meng F, Li R, Liu B. Combined delivery of salinomycin and docetaxel by dual-targeting gelatinase nanoparticles effectively inhibits cervical cancer cells and cancer stem cells. Drug Deliv 2021; 28:510-519. [PMID: 33657950 PMCID: PMC7935125 DOI: 10.1080/10717544.2021.1886378] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/02/2021] [Indexed: 01/05/2023] Open
Abstract
Intra-tumor heterogeneity is widely accepted as one of the key factors, which hinders cancer patients from achieving full recovery. Especially, cancer stem cells (CSCs) may exhibit self-renewal capacity, which makes it harder for complete elimination of tumor. Therefore, simultaneously inhibiting CSCs and non-CSCs in tumors becomes a promising strategy to obtain sustainable anticancer efficacy. Salinomycin (Sal) was reported to be critical to inhibit CSCs. However, the poor bioavailability and catastrophic side effects brought about limitations to clinical practice. To solve this problem, we previously constructed gelatinase-stimuli nanoparticles composed of nontoxic, biocompatible polyethylene glycol-polycaprolactone (PEG-PCL) copolymer with a gelatinase-cleavable peptide Pro-Val-Gly-Leu-Iso-Gly (PVGLIG) inserted between the two blocks of the copolymer. By applying our "smart" gelatinase-responsive nanoparticles for Sal delivery, we have demonstrated specific accumulation in tumor, anti-CSCs ability and reduced toxicity of Sal-NPs in our previous study. In the present study, we synthesized Sal-Docetaxel-loaded gelatinase-stimuli nanoparticles (Sal-Doc NP) and confirmed single emulsion as the optimal method of producing Sal-Doc NPs (Sal-Doc SE-NP) in comparison with nanoprecipitation. Sal-Doc SE-NPs inhibited both CSCs and non-CSCs in mice transplanted with cervical cancer, and might be associated with enhanced restriction of epithelial-mesenchymal transition (EMT) pathway. Besides, the tumorigenic capacity and growing speed were obviously suppressed in Sal-Doc-SE-NPs-treated group in rechallenge experiment. Our results suggest that Sal-Doc-loaded gelatinase-stimuli nanoparticles could be a promising strategy to enhance antitumor efficacy and reduce side effects by simultaneously suppressing CSCs and non-CSCs.
Collapse
Affiliation(s)
- Qin Wang
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Ying-Tzu Yen
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Chen Xie
- Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, China
| | - Fangcen Liu
- Department of pathology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qin Liu
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Jia Wei
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Lixia Yu
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Lifeng Wang
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Fanyan Meng
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Rutian Li
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Baorui Liu
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| |
Collapse
|
33
|
Qi D, Liu Y, Li J, Huang JH, Hu X, Wu E. Salinomycin as a potent anticancer stem cell agent: State of the art and future directions. Med Res Rev 2021; 42:1037-1063. [PMID: 34786735 PMCID: PMC9298915 DOI: 10.1002/med.21870] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 12/11/2022]
Abstract
Cancer stem cells (CSCs) are a small subpopulation of cells within a tumor that can both self‐renew and differentiate into other cell types forming the heterogeneous tumor bulk. Since CSCs are involved in all aspects of cancer development, including tumor initiation, cell proliferation, metastatic dissemination, therapy resistance, and recurrence, they have emerged as attractive targets for cancer treatment and management. Salinomycin, a widely used antibiotic in poultry farming, was identified by the Weinberg group as a potent anti‐CSC agent in 2009. As a polyether ionophore, salinomycin exerts broad‐spectrum activities, including the important anti‐CSC function. Studies on the mechanism of action of salinomycin against cancer have been continuously and rapidly published since then. Thus, it is imperative for us to update its literature of recent research findings in this area. We here summarize the notable work reported on salinomycin's anticancer activities, intracellular binding target(s), effects on tumor microenvironment, safety, derivatives, and tumor‐specific drug delivery; after that we also discuss the translational potential of salinomycin toward clinical application based on current multifaceted understandings.
Collapse
Affiliation(s)
- Dan Qi
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas, USA
| | - Yunyi Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China
| | - Juan Li
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China
| | - Jason H Huang
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas, USA.,Department of Surgery, Texas A&M University College of Medicine, Temple, Texas, USA
| | - Xiaoxiao Hu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China.,Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Erxi Wu
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas, USA.,Department of Surgery, Texas A&M University College of Medicine, Temple, Texas, USA.,LIVESTRONG Cancer Institutes and Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA.,Department of Pharmaceutical Sciences, Texas A&M University College of Pharmacy, College Station, Texas, USA
| |
Collapse
|
34
|
Jiang C, Zhou H, Sun H, He R, Song C, Cui T, Luan J, Fu J, Zhang Y, Jiao N, Wang H. Establishing an efficient salinomycin biosynthetic pathway in three heterologous Streptomyces hosts by constructing a 106-kb multioperon artificial gene cluster. Biotechnol Bioeng 2021; 118:4668-4677. [PMID: 34436784 DOI: 10.1002/bit.27928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/06/2021] [Accepted: 08/19/2021] [Indexed: 11/09/2022]
Abstract
Salinomycin is a promising anticancer drug for chemotherapy. A highly productive biosynthetic gene cluster will facilitate the creation of analogs with improved therapeutic activity and reduced side effects. In this study, we engineered an artificial 106-kb salinomycin gene cluster and achieved efficient heterologous expression in three hosts: Streptomyces coelicolor CH999, S. lividans K4-114, and S. albus J1074. The six-operon artificial gene cluster consists of 25 genes from the native gene cluster organized into five operons and five fatty acid β-oxidation genes into one operon. All operons are driven by strong constitutive promoters. For K4-114 and J1074 harboring the artificial gene cluster, salinomycin production in shake flask cultures was 14.3 mg L-1 and 19.3 mg L-1 , respectively. The production was 1.3-fold and 1.7-fold higher, respectively, than that of the native producer S. albus DSM41398. K4-114 and J1074 harboring the native gene cluster produced an undetectable amount of salinomycin and 0.5 mg L-1 , respectively. CH999 harboring the artificial gene cluster produced 10.3 mg L-1 of salinomycin, which was 92% of the production by DSM41398. The efficient heterologous expression system based on the 106-kb multioperon artificial gene cluster established in this study will facilitate structural diversification of salinomycin, which is valuable for drug development and structure-activity studies.
Collapse
Affiliation(s)
- Chanjuan Jiang
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, China.,State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Helmholtz International Lab for Anti-infectives, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, Shandong, China
| | - Haibo Zhou
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Helmholtz International Lab for Anti-infectives, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, Shandong, China
| | - Hongluan Sun
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Helmholtz International Lab for Anti-infectives, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, Shandong, China
| | - Ruoting He
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Helmholtz International Lab for Anti-infectives, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, Shandong, China
| | - Chaoyi Song
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Helmholtz International Lab for Anti-infectives, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, Shandong, China
| | - Tianqi Cui
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Helmholtz International Lab for Anti-infectives, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, Shandong, China
| | - Ji Luan
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Helmholtz International Lab for Anti-infectives, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, Shandong, China
| | - Jun Fu
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Helmholtz International Lab for Anti-infectives, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, Shandong, China
| | - Youming Zhang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Helmholtz International Lab for Anti-infectives, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, Shandong, China
| | - Nianzhi Jiao
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, China
| | - Hailong Wang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Helmholtz International Lab for Anti-infectives, Shandong University-Helmholtz Institute of Biotechnology, Shandong University, Qingdao, Shandong, China
| |
Collapse
|
35
|
Rodriguez D, Watts D, Gaete D, Sormendi S, Wielockx B. Hypoxia Pathway Proteins and Their Impact on the Blood Vasculature. Int J Mol Sci 2021; 22:ijms22179191. [PMID: 34502102 PMCID: PMC8431527 DOI: 10.3390/ijms22179191] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/16/2021] [Accepted: 08/21/2021] [Indexed: 12/12/2022] Open
Abstract
Every cell in the body requires oxygen for its functioning, in virtually every animal, and a tightly regulated system that balances oxygen supply and demand is therefore fundamental. The vascular network is one of the first systems to sense oxygen, and deprived oxygen (hypoxia) conditions automatically lead to a cascade of cellular signals that serve to circumvent the negative effects of hypoxia, such as angiogenesis associated with inflammation, tumor development, or vascular disorders. This vascular signaling is driven by central transcription factors, namely the hypoxia inducible factors (HIFs), which determine the expression of a growing number of genes in endothelial cells and pericytes. HIF functions are tightly regulated by oxygen sensors known as the HIF-prolyl hydroxylase domain proteins (PHDs), which are enzymes that hydroxylate HIFs for eventual proteasomal degradation. HIFs, as well as PHDs, represent attractive therapeutic targets under various pathological settings, including those involving vascular (dys)function. We focus on the characteristics and mechanisms by which vascular cells respond to hypoxia under a variety of conditions.
Collapse
|
36
|
Roudsari NM, Lashgari NA, Momtaz S, Abaft S, Jamali F, Safaiepour P, Narimisa K, Jackson G, Bishayee A, Rezaei N, Abdolghaffari AH, Bishayee A. Inhibitors of the PI3K/Akt/mTOR Pathway in Prostate Cancer Chemoprevention and Intervention. Pharmaceutics 2021; 13:1195. [PMID: 34452154 PMCID: PMC8400324 DOI: 10.3390/pharmaceutics13081195] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/serine-threonine kinase (Akt)/mammalian target of the rapamycin (mTOR)-signaling pathway has been suggested to have connections with the malignant transformation, growth, proliferation, and metastasis of various cancers and solid tumors. Relevant connections between the PI3K/Akt/mTOR pathway, cell survival, and prostate cancer (PC) provide a great therapeutic target for PC prevention or treatment. Recent studies have focused on small-molecule mTOR inhibitors or their usage in coordination with other therapeutics for PC treatment that are currently undergoing clinical testing. In this study, the function of the PI3K/Akt/mTOR pathway, the consequence of its dysregulation, and the development of mTOR inhibitors, either as an individual substance or in combination with other agents, and their clinical implications are discussed. The rationale for targeting the PI3K/Akt/mTOR pathway, and specifically the application and potential utility of natural agents involved in PC treatment is described. In addition to the small-molecule mTOR inhibitors, there are evidence that several natural agents are able to target the PI3K/Akt/mTOR pathway in prostatic neoplasms. These natural mTOR inhibitors can interfere with the PI3K/Akt/mTOR pathway through multiple mechanisms; however, inhibition of Akt and suppression of mTOR 1 activity are two major therapeutic approaches. Combination therapy improves the efficacy of these inhibitors to either suppress the PC progression or circumvent the resistance by cancer cells.
Collapse
Affiliation(s)
- Nazanin Momeni Roudsari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran 1941933111, Iran; (N.M.R.); (N.-A.L.); (S.A.); (F.J.); (P.S.); (K.N.)
| | - Naser-Aldin Lashgari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran 1941933111, Iran; (N.M.R.); (N.-A.L.); (S.A.); (F.J.); (P.S.); (K.N.)
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, Academic Center for Education, Culture and Research, Tehran 1417614411, Iran;
- Toxicology and Disease Group, Pharmaceutical Sciences Research Center, Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
- Gastrointestinal Pharmacology Interest Group, Universal Scientific Education and Research Network, Tehran 1417614411, Iran
| | - Shaghayegh Abaft
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran 1941933111, Iran; (N.M.R.); (N.-A.L.); (S.A.); (F.J.); (P.S.); (K.N.)
| | - Fatemeh Jamali
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran 1941933111, Iran; (N.M.R.); (N.-A.L.); (S.A.); (F.J.); (P.S.); (K.N.)
| | - Pardis Safaiepour
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran 1941933111, Iran; (N.M.R.); (N.-A.L.); (S.A.); (F.J.); (P.S.); (K.N.)
| | - Kiyana Narimisa
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran 1941933111, Iran; (N.M.R.); (N.-A.L.); (S.A.); (F.J.); (P.S.); (K.N.)
| | - Gloria Jackson
- Lake Erie Collage of Osteopathic Medicine, Bradenton, FL 34211, USA;
| | | | - Nima Rezaei
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran 1417614411, Iran;
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Amir Hossein Abdolghaffari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran 1941933111, Iran; (N.M.R.); (N.-A.L.); (S.A.); (F.J.); (P.S.); (K.N.)
- Medicinal Plants Research Center, Institute of Medicinal Plants, Academic Center for Education, Culture and Research, Tehran 1417614411, Iran;
- Toxicology and Disease Group, Pharmaceutical Sciences Research Center, Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
- Gastrointestinal Pharmacology Interest Group, Universal Scientific Education and Research Network, Tehran 1417614411, Iran
| | - Anupam Bishayee
- Lake Erie Collage of Osteopathic Medicine, Bradenton, FL 34211, USA;
| |
Collapse
|
37
|
Shen H, Chen Y, Wan Y, Liu T, Wang J, Zhang Y, Wei L, Hu Q, Xu B, Chernov M, Frangou C, Zhang J. Identification of TAZ-Dependent Breast Cancer Vulnerabilities Using a Chemical Genomics Screening Approach. Front Cell Dev Biol 2021; 9:673374. [PMID: 34211974 PMCID: PMC8239392 DOI: 10.3389/fcell.2021.673374] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/19/2021] [Indexed: 12/22/2022] Open
Abstract
Breast cancer stem cells (BCSCs) represent a subpopulation of tumor cells that can self-renew and generate tumor heterogeneity. Targeting BCSCs may ameliorate therapy resistance, tumor growth, and metastatic progression. However, the origin and molecular mechanisms underlying their cellular properties are poorly understood. The transcriptional coactivator with PDZ-binding motif (TAZ) promotes mammary stem/progenitor cell (MaSC) expansion and maintenance but also confers stem-like traits to differentiated tumor cells. Here, we describe the rapid generation of experimentally induced BCSCs by TAZ-mediated reprogramming of human mammary epithelial cells, hence allowing for the direct analysis of BCSC phenotypes. Specifically, we establish genetically well-defined TAZ-dependent (TAZDEP) and -independent (TAZIND) cell lines with cancer stem cell (CSC) traits, such as self-renewal, variable resistance to chemotherapeutic agents, and tumor seeding potential. TAZDEP cells were associated with the epithelial to mesenchymal transition, embryonic, and MaSC signature genes. In contrast, TAZIND cells were characterized by a neuroendocrine transdifferentiation transcriptional program associated with Polycomb repressive complex 2 (PRC2). Mechanistically, we identify Cyclin D1 (CCND1) as a critical downstream effector for TAZ-driven tumorigenesis. Overall, our results reveal a critical TAZ-CCND1-CDK4/CDK6 signaling axis, suggesting novel therapeutic approaches to eliminate both BCSCs and therapy-resistant cancer cells.
Collapse
Affiliation(s)
- He Shen
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Yanmin Chen
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Yin Wan
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Tao Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Yali Zhang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Lei Wei
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Qiang Hu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Bo Xu
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Mikhail Chernov
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Costa Frangou
- Molecular and Integrative Physiology Department, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Jianmin Zhang
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
38
|
Asik A, Ay NPO, Bagca BG, Caglar HO, Gunduz C, Avci CB. Combination of Salinomycin and AZD3463 Reveals Synergistic Effect on Reducing the Viability of T98G Glioblastoma Cells. Anticancer Agents Med Chem 2021; 20:2267-2273. [PMID: 32698744 DOI: 10.2174/1871520620666200721121517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Salinomycin, an ionophore antibiotic, is known to be an effective agent in reducing the viability of Glioblastoma (GBM) cells. The combination of salinomycin with other chemotherapeutic drugs would help to overcome the drug resistance of GBM cells. OBJECTIVE This study aims to test the combinatorial effect of salinomycin and AZD3463 in T98G GBM cells. METHODS The cytotoxic effects of drugs on T98G GBM cells were determined by using WST-8 assay. Flow cytometry was used to identify apoptosis and cell cycle profiles after treatments. Real-time PCR was used to portray mRNA expression profiles of genes in the Wnt-signaling pathway after treatments. RESULTS IC50 concentrations of AZD3463 and salinomycin were 529nM and 7.3μM for 48h, respectively. The combination concentrations of AZD3463 and salinomycin were 3.3μM and 333nM, respectively. The combination treatment showed a synergistic effect on reducing the viability of GBM cells. AZD3463, salinomycin, and their combination induced apoptosis in 1.2, 1.4, and 3.2 folds, respectively. AZD3463 and the combination treatment induced the cell cycle arrest at the G1 phase. Salinomycin and AZD3463 treatments, either alone or in combination, resulted in the downregulation or upregulation of mRNA expression levels of genes in the Wntsignaling pathway. CONCLUSION Salinomycin, AZD3463, and their combination may inhibit proliferation and induce apoptosis in GBM cells due to a decrease in expression levels of genes acting in both the canonical and non-canonical Wnt signaling pathways. The Wnt signaling pathway may be involved in salinomycin-AZD3463 drug interaction.
Collapse
Affiliation(s)
- Aycan Asik
- Department of Medical Biology, Medical Faculty, Ege University, Bornova, 35100, Izmir, Turkey
| | - Neslihan P O Ay
- Department of Medical Biology, Medical Faculty, Ege University, Bornova, 35100, Izmir, Turkey
| | - Bakiye G Bagca
- Department of Medical Biology, Medical Faculty, Ege University, Bornova, 35100, Izmir, Turkey
| | - Hasan O Caglar
- Department of Stem Cell, Health Science Institute, Ege University, Bornova, 35100, Izmir, Turkey
| | - Cumhur Gunduz
- Department of Medical Biology, Medical Faculty, Ege University, Bornova, 35100, Izmir, Turkey
| | - Cigir B Avci
- Department of Medical Biology, Medical Faculty, Ege University, Bornova, 35100, Izmir, Turkey
| |
Collapse
|
39
|
Burke RM, Dirkx RA, Quijada P, Lighthouse JK, Mohan A, O'Brien M, Wojciechowski W, Woeller CF, Phipps RP, Alexis JD, Ashton JM, Small EM. Prevention of Fibrosis and Pathological Cardiac Remodeling by Salinomycin. Circ Res 2021; 128:1663-1678. [PMID: 33825488 DOI: 10.1161/circresaha.120.317791] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Ryan M Burke
- Aab Cardiovascular Research Institute, Department of Medicine (R.M.B., R.A.D., P.Q., J.K.L., A.M., E.M.S.), University of Rochester School of Medicine and Dentistry, NY
| | - Ronald A Dirkx
- Aab Cardiovascular Research Institute, Department of Medicine (R.M.B., R.A.D., P.Q., J.K.L., A.M., E.M.S.), University of Rochester School of Medicine and Dentistry, NY
| | - Pearl Quijada
- Aab Cardiovascular Research Institute, Department of Medicine (R.M.B., R.A.D., P.Q., J.K.L., A.M., E.M.S.), University of Rochester School of Medicine and Dentistry, NY
| | - Janet K Lighthouse
- Aab Cardiovascular Research Institute, Department of Medicine (R.M.B., R.A.D., P.Q., J.K.L., A.M., E.M.S.), University of Rochester School of Medicine and Dentistry, NY
| | - Amy Mohan
- Aab Cardiovascular Research Institute, Department of Medicine (R.M.B., R.A.D., P.Q., J.K.L., A.M., E.M.S.), University of Rochester School of Medicine and Dentistry, NY
| | - Meghann O'Brien
- Genomics Research Center (M.O., W.W., J.M.A.), University of Rochester School of Medicine and Dentistry, NY
| | - Wojciech Wojciechowski
- Genomics Research Center (M.O., W.W., J.M.A.), University of Rochester School of Medicine and Dentistry, NY
| | - Collynn F Woeller
- Environmental Medicine (C.F.W., R.P.P.), University of Rochester School of Medicine and Dentistry, NY.,Department of Medicine (C.F.W., R.P.P., J.D.A., E.M.S.), University of Rochester School of Medicine and Dentistry, NY
| | - Richard P Phipps
- Environmental Medicine (C.F.W., R.P.P.), University of Rochester School of Medicine and Dentistry, NY.,Department of Medicine (C.F.W., R.P.P., J.D.A., E.M.S.), University of Rochester School of Medicine and Dentistry, NY
| | - Jeffrey D Alexis
- Department of Medicine (C.F.W., R.P.P., J.D.A., E.M.S.), University of Rochester School of Medicine and Dentistry, NY
| | - John M Ashton
- Genomics Research Center (M.O., W.W., J.M.A.), University of Rochester School of Medicine and Dentistry, NY
| | - Eric M Small
- Aab Cardiovascular Research Institute, Department of Medicine (R.M.B., R.A.D., P.Q., J.K.L., A.M., E.M.S.), University of Rochester School of Medicine and Dentistry, NY.,Department of Medicine (C.F.W., R.P.P., J.D.A., E.M.S.), University of Rochester School of Medicine and Dentistry, NY.,Pharmacology and Physiology (E.M.S.), University of Rochester School of Medicine and Dentistry, NY.,Biomedical Engineering, University of Rochester, NY (E.M.S.)
| |
Collapse
|
40
|
Yuan C, Huang X, Zhai R, Ma Y, Xu A, Zhang P, Yang Q. In Vitro Antiviral Activities of Salinomycin on Porcine Epidemic Diarrhea Virus. Viruses 2021; 13:v13040580. [PMID: 33808275 PMCID: PMC8066218 DOI: 10.3390/v13040580] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/25/2021] [Accepted: 03/28/2021] [Indexed: 01/16/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV), an enteropathogenic coronavirus, has catastrophic impacts on the global pig industry. Owing to the lack of effective vaccines and specific therapeutic options for PEDV, it is pertinent to develop new and available antivirals. This study identified, for the first time, a salinomycin that actively inhibited PEDV replication in Vero cells in a dose-dependent manner. Furthermore, salinomycin significantly inhibited PEDV infection by suppressing the entry and post-entry of PEDV in Vero cells. It did not directly interact with or inactivate PEDV particles, but it significantly ameliorated the activation of Erk1/2, JNK and p38MAPK signaling pathways that are associated with PEDV infection. This implied that salinomycin inhibits PEDV replication by altering MAPK pathway activation. Notably, the PEDV induced increase in reactive oxidative species (ROS) was not decreased, indicating that salinomycin suppresses PEDV replication through a pathway that is an independent pathway of viral-induced ROS. Therefore, salinomycin is a potential drug that can be used for treating PEDV infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qian Yang
- Correspondence: ; Tel.: +86-02584395817
| |
Collapse
|
41
|
Suresh P, Thamotharan S, Selva Ganesan S. NHC Organocatalysis in D
2
O for the Highly Diastereoselective Synthesis of Deuterated Spiropyran Analogues. ChemistrySelect 2021. [DOI: 10.1002/slct.202004150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Pavithira Suresh
- Department of Chemistry, School of Chemical and Biotechnology SASTRA Deemed University Thanjavur- 613401 Tamil Nadu India
| | - Subbiah Thamotharan
- Biomolecular Crystallography Laboratory, Department of Bioinformatics, School of Chemical and Biotechnology SASTRA Deemed University Thanjavur- 613401 Tamil Nadu India
| | - Subramaniapillai Selva Ganesan
- Department of Chemistry, School of Chemical and Biotechnology SASTRA Deemed University Thanjavur- 613401 Tamil Nadu India
| |
Collapse
|
42
|
Valdivia G, Alonso-Diez Á, Pérez-Alenza D, Peña L. From Conventional to Precision Therapy in Canine Mammary Cancer: A Comprehensive Review. Front Vet Sci 2021; 8:623800. [PMID: 33681329 PMCID: PMC7925635 DOI: 10.3389/fvets.2021.623800] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
Canine mammary tumors (CMTs) are the most common neoplasm in intact female dogs. Canine mammary cancer (CMC) represents 50% of CMTs, and besides surgery, which is the elective treatment, additional targeted and non-targeted therapies could offer benefits in terms of survival to these patients. Also, CMC is considered a good spontaneous intermediate animal model for the research of human breast cancer (HBC), and therefore, the study of new treatments for CMC is a promising field in comparative oncology. Dogs with CMC have a comparable disease, an intact immune system, and a much shorter life span, which allows the achievement of results in a relatively short time. Besides conventional chemotherapy, innovative therapies have a large niche of opportunities. In this article, a comprehensive review of the current research in adjuvant therapies for CMC is conducted to gather available information and evaluate the perspectives. Firstly, updates are provided on the clinical-pathological approach and the use of conventional therapies, to delve later into precision therapies against therapeutic targets such as hormone receptors, tyrosine kinase receptors, p53 tumor suppressor gene, cyclooxygenases, the signaling pathways involved in epithelial-mesenchymal transition, and immunotherapy in different approaches. A comparison of the different investigations on targeted therapies in HBC is also carried out. In the last years, the increasing number of basic research studies of new promising therapeutic agents on CMC cell lines and CMC mouse xenografts is outstanding. As the main conclusion of this review, the lack of effort to bring the in vitro studies into the field of applied clinical research emerges. There is a great need for well-planned large prospective randomized clinical trials in dogs with CMC to obtain valid results for both species, humans and dogs, on the use of new therapies. Following the One Health concept, human and veterinary oncology will have to join forces to take advantage of both the economic and technological resources that are invested in HBC research, together with the innumerable advantages of dogs with CMC as a spontaneous animal model.
Collapse
Affiliation(s)
- Guillermo Valdivia
- Department Animal Medicine, Surgery and Pathology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Mammary Oncology Unit, Complutense Veterinary Teaching Hospital, Complutense University of Madrid, Madrid, Spain
| | - Ángela Alonso-Diez
- Department Animal Medicine, Surgery and Pathology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Mammary Oncology Unit, Complutense Veterinary Teaching Hospital, Complutense University of Madrid, Madrid, Spain
| | - Dolores Pérez-Alenza
- Department Animal Medicine, Surgery and Pathology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Mammary Oncology Unit, Complutense Veterinary Teaching Hospital, Complutense University of Madrid, Madrid, Spain
| | - Laura Peña
- Department Animal Medicine, Surgery and Pathology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Mammary Oncology Unit, Complutense Veterinary Teaching Hospital, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
43
|
Trailblazing perspectives on targeting breast cancer stem cells. Pharmacol Ther 2021; 223:107800. [PMID: 33421449 DOI: 10.1016/j.pharmthera.2021.107800] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Breast cancer (BCa) is one of the most prevalent malignant tumors affecting women's health worldwide. The recurrence and metastasis of BCa have made it a long-standing challenge to achieve remission-persistent or disease-undetectable clinical outcomes. Cancer stem cells (CSCs) possess the ability to self-renew and generate heterogeneous tumor bulk. The existence of CSCs has been found to be vital in the initiation, metastasis, therapy resistance, and recurrence of tumors across cancer types. Because CSCs grow slowly in their dormant state, they are insensitive to conventional chemotherapies; however, when CSCs emerge from their dormant state and become clinically evident, they usually acquire genetic traits that make them resistant to existing therapies. Moreover, CSCs also show evidence of acquired drug resistance in synchrony with tumor relapses. The concept of CSCs provides a new treatment strategy for BCa. In this review, we highlight the recent advances in research on breast CSCs and their association with epithelial-mesenchymal transition (EMT), circulating tumor cells (CTCs), plasticity of tumor cells, tumor microenvironment (TME), T-cell modulatory protein PD-L1, and non-coding RNAs. On the basis that CSCs are associated with multiple dysregulated biological processes, we envisage that increased understanding of disease sub-classification, selected combination of conventional treatment, molecular aberration directed therapy, immunotherapy, and CSC targeting/sensitizing strategy might improve the treatment outcome of patients with advanced BCa. We also discuss novel perspectives on new drugs and therapeutics purposing the potent and selective expunging of CSCs.
Collapse
|
44
|
Kozak J, Forma A, Czeczelewski M, Kozyra P, Sitarz E, Radzikowska-Büchner E, Sitarz M, Baj J. Inhibition or Reversal of the Epithelial-Mesenchymal Transition in Gastric Cancer: Pharmacological Approaches. Int J Mol Sci 2020; 22:ijms22010277. [PMID: 33383973 PMCID: PMC7795012 DOI: 10.3390/ijms22010277] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) constitutes one of the hallmarks of carcinogenesis consisting in the re-differentiation of the epithelial cells into mesenchymal ones changing the cellular phenotype into a malignant one. EMT has been shown to play a role in the malignant transformation and while occurring in the tumor microenvironment, it significantly affects the aggressiveness of gastric cancer, among others. Importantly, after EMT occurs, gastric cancer patients are more susceptible to the induction of resistance to various therapeutic agents, worsening the clinical outcome of patients. Therefore, there is an urgent need to search for the newest pharmacological agents targeting EMT to prevent further progression of gastric carcinogenesis and potential metastases. Therapies targeted at EMT might be combined with other currently available treatment modalities, which seems to be an effective strategy to treat gastric cancer patients. In this review, we have summarized recent advances in gastric cancer treatment in terms of targeting EMT specifically, such as the administration of polyphenols, resveratrol, tangeretin, luteolin, genistein, proton pump inhibitors, terpenes, other plant extracts, or inorganic compounds.
Collapse
Affiliation(s)
- Joanna Kozak
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Alicja Forma
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (A.F.); (M.C.)
| | - Marcin Czeczelewski
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (A.F.); (M.C.)
| | - Paweł Kozyra
- Student Research Group, Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, PL-20093 Lublin, Poland;
| | - Elżbieta Sitarz
- 1st Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, Gluska Street 1, 20-439 Lublin, Poland;
| | - Elżbieta Radzikowska-Büchner
- Department of Plastic Surgery, Central Clinical Hospital of the Ministry of the Interior in Warsaw, 01-211 Warsaw, Poland;
| | - Monika Sitarz
- Department of Conservative Dentistry with Endodontics, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Jacek Baj
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland;
- Correspondence:
| |
Collapse
|
45
|
Heffer AM, Wang V, Libby RT, Feldon SE, Woeller CF, Kuriyan AE. Salinomycin inhibits proliferative vitreoretinopathy formation in a mouse model. PLoS One 2020; 15:e0243626. [PMID: 33347461 PMCID: PMC7751870 DOI: 10.1371/journal.pone.0243626] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/24/2020] [Indexed: 01/22/2023] Open
Abstract
Proliferative vitreoretinopathy (PVR) is a progressive disease that develops in a subset of patients who undergo surgery for retinal detachment repair, and results in significant vision loss. PVR is characterized by the migration of retinal pigment epithelial (RPE) cells into the vitreous cavity, where they undergo epithelial-to-mesenchymal transition and form contractile membranes within the vitreous and along the retina, resulting in recurrent retinal detachments. Currently, surgical intervention is the only treatment for PVR and there are no pharmacological agents that effectively inhibit or prevent PVR formation. Here, we show that a single intravitreal injection of the polyether ionophore salinomycin (SNC) effectively inhibits the formation of PVR in a mouse model with no evidence of retinal toxicity. After 4 weeks, fundus photography and optical coherence tomography (OCT) demonstrated development of mean PVR grade of 3.5 (SD: 1.3) in mouse eyes injected with RPE cells/DMSO (vehicle), compared to mean PVR grade of 1.6 (SD: 1.3) in eyes injected with RPE cells/SNC (p = 0.001). Additionally, immunohistochemistry analysis showed RPE cells/SNC treatment reduced both fibrotic (αSMA, FN1, Vim) and inflammatory (GFAP, CD3, CD20) markers compared to control RPE cells/DMSO treatment. Finally, qPCR analysis confirmed that Tgfβ, Tnfα, Mcp1 (inflammatory/cytokine markers), and Fn1, Col1a1 and Acta2 (fibrotic markers) were significantly attenuated in the RPE cells/SNC group compared to RPE/DMSO control. These results suggest that SNC is a potential pharmacologic agent for the prevention of PVR in humans and warrants further investigation.
Collapse
Affiliation(s)
- Alison M. Heffer
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
| | - Victor Wang
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
| | - Richard T. Libby
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
- Center for Visual Sciences, University of Rochester, Rochester, NY, United States of America
| | - Steven E. Feldon
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
- Center for Visual Sciences, University of Rochester, Rochester, NY, United States of America
| | - Collynn F. Woeller
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
| | - Ajay E. Kuriyan
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
- Center for Visual Sciences, University of Rochester, Rochester, NY, United States of America
- Retina Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, United States of America
| |
Collapse
|
46
|
Ebokaiwe AP, Njoya EM, Sheng Y, Zhang Z, Li S, Zhou Z, Qiang Z, Peng T, Hussein AA, Zhang G, Lu X, Li L, Wang F. Salinomycin promotes T-cell proliferation by inhibiting the expression and enzymatic activity of immunosuppressive indoleamine-2,3-dioxygenase in human breast cancer cells. Toxicol Appl Pharmacol 2020; 404:115203. [PMID: 32822738 DOI: 10.1016/j.taap.2020.115203] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 01/25/2023]
Abstract
Indoleamine 2,3 dioxygenase (IDO) is upregulated in many tumor types, including breast cancer, and plays a reputable role in promoting tumor immune tolerance. The importance of the immunosuppressive mechanism of IDO by suppressing T-cell function has garnered profound interest in the development of clinical IDO inhibitors. Herein, we established a screening method with cervical HeLa cells to induce IDO expression using interferon-γ (IFN-γ). After screening our chemical library, we found that salinomycin potently inhibited IFN-γ-stimulated kynurenine synthesis with IC50 values of 3.36-4.66 μM in both human cervical and breast cancer cells. Salinomycin lowered the IDO1 and IDO2 expression with no impact on the expression of tryptophan-2,3-dioxygenase. Interestingly, salinomycin potently repressed the IDO1 enzymatic activity by directly targeting the proteins in cells. Molecular docking revealed an alignment that favors nucleophilic attack of salinomycin in the catalytic domain of IDO1. Activation of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway by IFN-γ was significantly suppressed by salinomycin, via inhibiting the Jak1, Jak2, and STAT1/3 phosphorylation. Moreover, it inhibited IFN-γ-induced activation of the nuclear factor (NF)-κB pathway by inhibiting IκB degradation and NF-κB phosphorylation without affecting BIN1 expression. Furthermore, salinomycin significantly restored the proliferation of T cells co-cultured with IFN-γ-treated breast cancer cells and potentiated antitumor activity of cisplatin in vivo. These findings suggest that salinomycin suppresses kynurenine synthesis by inhibiting the catalytic activity of IDO1 and its expression by inhibiting the JAK/STAT and NF-κB pathways. Salinomycin warrants further investigation as a novel dual-functional IDO inhibitor for cancer immunotherapy.
Collapse
Affiliation(s)
- Azubuike Peter Ebokaiwe
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; Department of Chemistry/Biochemistry and Molecular Biology, Alex Ekwueme Federal University Ndufu Alike-, Ikwo, Nigeria
| | - Emmanuel Mfotie Njoya
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; Department of Biochemistry, Faculty of Science, University of Yaoundé I, P.O Box 812, Yaoundé, Cameroon
| | - Yuwen Sheng
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Zhonghui Zhang
- College of Chemical Engineering, Sichuan University, Chengdu 610064, China
| | - Sheng Li
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Zongyuan Zhou
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Zhe Qiang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Ting Peng
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Ahmed A Hussein
- Chemistry Department, Cape Peninsula University of Technology, Bellville Campus, Bellville 7537, Western Cape, South Africa
| | - Guolin Zhang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Xiaoxia Lu
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Lin Li
- Department of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, China.
| | - Fei Wang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China.
| |
Collapse
|
47
|
Jayawardhana AMDS, Stilgenbauer M, Datta P, Qiu Z, Mckenzie S, Wang H, Bowers D, Kurokawa M, Zheng YR. Fatty acid-like Pt(IV) prodrugs overcome cisplatin resistance in ovarian cancer by harnessing CD36. Chem Commun (Camb) 2020; 56:10706-10709. [PMID: 32789350 DOI: 10.1039/d0cc02174a] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Resistance to the platinum-based chemotherapy drug, cisplatin, is a significant setback in ovarian cancer. We engineered fatty acid-like Pt(iv) prodrugs that harness the fatty acid transporter CD36 to facilitate their entry to ovarian cancer cells. We show that these novel constructs effectively kill cisplatin-resistant ovarian cancer cells.
Collapse
|
48
|
Su Y, Zhao B, Zhou L, Zhang Z, Shen Y, Lv H, AlQudsy LHH, Shang P. Ferroptosis, a novel pharmacological mechanism of anti-cancer drugs. Cancer Lett 2020; 483:127-136. [DOI: 10.1016/j.canlet.2020.02.015] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/12/2020] [Accepted: 02/12/2020] [Indexed: 02/06/2023]
|
49
|
Fan S, Price T, Huang W, Plue M, Warren J, Sundaramoorthy P, Paul B, Feinberg D, MacIver N, Chao N, Sipkins D, Kang Y. PINK1-Dependent Mitophagy Regulates the Migration and Homing of Multiple Myeloma Cells via the MOB1B-Mediated Hippo-YAP/TAZ Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1900860. [PMID: 32154065 PMCID: PMC7055555 DOI: 10.1002/advs.201900860] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 11/23/2019] [Indexed: 05/07/2023]
Abstract
The roles of mitochondrial dysfunction in carcinogenesis remain largely unknown. The effects of PTEN-induced putative kinase 1 (PINK1)-dependent mitophagy on the pathogenesis of multiple myeloma (MM) are determined. The levels of the PINK1-dependent mitophagy markers PINK1 and parkin RBR E3 ubiquitin protein ligase (PARK2) in CD138+ plasma cells are reduced in patients with MM and correlate with clinical outcomes in myeloma patients. Moreover, the induction of PINK1-dependent mitophagy with carbonylcyanide-m-chlorophenylhydrazone (CCCP) or salinomycin, or overexpression of PINK1 leads to inhibition of transwell migration, suppression of myeloma cell homing to calvarium, and decreased osteolytic bone lesions. Furthermore, genetic deletion of pink1 accelerates myeloma development in a spontaneous X-box binding protein-1 spliced isoform (XBP-1s) transgenic myeloma mouse model and in VK*MYC transplantable myeloma recipient mice. Additionally, treatment with salinomycin shows significant antimyeloma activities in vivo in murine myeloma xenograft models. Finally, the effects of PINK1-dependent mitophagy on myeloma pathogenesis are driven by the activation of the Mps one binder kinase activator (MOB1B)-mediated Hippo pathway and the subsequent downregulation of Yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif (TAZ) expression. These data provide direct evidence that PINK1-dependent mitophagy plays a critical role in the pathogenesis of MM and is a potential therapeutic target.
Collapse
Affiliation(s)
- Shengjun Fan
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | - Trevor Price
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | - Wei Huang
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | - Michelle Plue
- Shared Materials Instrumentation FacilityPratt School of EngineeringDuke UniversityDurhamNC27708USA
| | | | - Pasupathi Sundaramoorthy
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | - Barry Paul
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | - Daniel Feinberg
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | | | - Nelson Chao
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | - Dorothy Sipkins
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| |
Collapse
|
50
|
Synthesis of chemical tools to improve water solubility and promote the delivery of salinomycin to cancer cells. Exp Ther Med 2020; 19:1835-1843. [PMID: 32104239 PMCID: PMC7027072 DOI: 10.3892/etm.2019.8368] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy and radiation are unable to eliminate all cancer cells, particularly apoptosis-resistant cancer cells, despite their ability to kill cancer cluster cells. Thus, it is important to identify methods that eliminate all cancer cells in order to prevent relapse. Salinomycin has the ability to control and eradicate different types of cancer, including breast cancer; however, its molecular mechanism remains unclear. The main difficulty in testing salinomycin activity and understanding the governing mechanisms is its low solubility in water (17 mg/l), which can hinder convenient delivery of salinomycin to the protein receptor at the cell surface of stem cells. In the present study, salinomycin was conjugated to the trans-activator of transcription-protein in order to facilitate its delivery to the cancer cells. Conjugated salinomycin demonstrated improved solubility in both in vitro. Salinomycin was tested in breast cancer cells (MCF7 and JIMT-1) by the cleavage of the linker through photolysis at l≥365 nm during in vitro analysis, in the present study.
Collapse
|