1
|
Hashim GM, Shahgolzari M, Hefferon K, Yavari A, Venkataraman S. Plant-Derived Anti-Cancer Therapeutics and Biopharmaceuticals. Bioengineering (Basel) 2024; 12:7. [PMID: 39851281 PMCID: PMC11759177 DOI: 10.3390/bioengineering12010007] [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: 11/13/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/26/2025] Open
Abstract
In spite of significant advancements in diagnosis and treatment, cancer remains one of the major threats to human health due to its ability to cause disease with high morbidity and mortality. A multifactorial and multitargeted approach is required towards intervention of the multitude of signaling pathways associated with carcinogenesis inclusive of angiogenesis and metastasis. In this context, plants provide an immense source of phytotherapeutics that show great promise as anticancer drugs. There is increasing epidemiological data indicating that diets rich in vegetables and fruits could decrease the risks of certain cancers. Several studies have proved that natural plant polyphenols, such as flavonoids, lignans, phenolic acids, alkaloids, phenylpropanoids, isoprenoids, terpenes, and stilbenes, could be used in anticancer prophylaxis and therapeutics by recruitment of mechanisms inclusive of antioxidant and anti-inflammatory activities and modulation of several molecular events associated with carcinogenesis. The current review discusses the anticancer activities of principal phytochemicals with focus on signaling circuits towards targeted cancer prophylaxis and therapy. Also addressed are plant-derived anti-cancer vaccines, nanoparticles, monoclonal antibodies, and immunotherapies. This review article brings to light the importance of plants and plant-based platforms as invaluable, low-cost sources of anti-cancer molecules of particular applicability in resource-poor developing countries.
Collapse
Affiliation(s)
- Ghyda Murad Hashim
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada
| | - Mehdi Shahgolzari
- Dental Research Center, Hamadan University of Medical Sciences, Hamadan 65175-4171, Iran
| | - Kathleen Hefferon
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada
| | - Afagh Yavari
- Department of Biology, Payame Noor University, Tehran P.O. Box 19395-3697, Iran
| | - Srividhya Venkataraman
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada
| |
Collapse
|
2
|
Fontana F, Molinaro G, Moroni S, Pallozzi G, Ferreira MPA, Tello RP, Elbadri K, Torrieri G, Correia A, Kemell M, Casettari L, Celia C, Santos HA. Biomimetic Platelet-Cloaked Nanoparticles for the Delivery of Anti-Inflammatory Curcumin in the Treatment of Atherosclerosis. Adv Healthc Mater 2024; 13:e2302074. [PMID: 38499190 PMCID: PMC11468963 DOI: 10.1002/adhm.202302074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 03/12/2024] [Indexed: 03/20/2024]
Abstract
Atherosclerosis still represents a major driver of cardiovascular diseases worldwide. Together with accumulation of lipids in the plaque, inflammation is recognized as one of the key players in the formation and development of atherosclerotic plaque. Systemic anti-inflammatory treatments are successful in reducing the disease burden, but are correlated with severe side effects, underlining the need for targeted formulations. In this work, curcumin is chosen as the anti-inflammatory payload model and further loaded in lignin-based nanoparticles (NPs). The NPs are then coated with a tannic acid (TA)- Fe (III) complex and further cloaked with fragments derived from platelet cell membrane, yielding NPs with homogenous size. The two coatings increase the interaction between the NPs and cells, both endothelial and macrophages, in steady state or inflamed status. Furthermore, NPs are cytocompatible toward endothelial, smooth muscle and immune cells, while not inducing immune activation. The anti-inflammatory efficacy is demonstrated in endothelial cells by real-time quantitative polymerase chain reaction and ELISA assay where curcumin-loaded NPs decrease the expression of Nf-κb, TGF-β1, IL-6, and IL-1β in lipopolysaccharide-inflamed cells. Overall, due to the increase in the cell-NP interactions and the anti-inflammatory efficacy, these NPs represent potential candidates for the targeted anti-inflammatory treatment of atherosclerosis.
Collapse
Affiliation(s)
- Flavia Fontana
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Giuseppina Molinaro
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Sofia Moroni
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Department of Biomolecular SciencesSchool of PharmacyUniversity of Urbino Carlo BoUrbinoI‐61029Italy
| | - Giulia Pallozzi
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Department of PharmacyUniversity of Chieti‐Pescara “G. D'Annunzio”Via dei Vestini 13ChietiI‐66100Italy
| | - Mónica P. A. Ferreira
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Present address:
MedEngine OyEteläranta 14Helsinki00130Finland
| | - Rubén Pareja Tello
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Khalil Elbadri
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Giulia Torrieri
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Alexandra Correia
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Marianna Kemell
- Department of ChemistryUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Luca Casettari
- Department of Biomolecular SciencesSchool of PharmacyUniversity of Urbino Carlo BoUrbinoI‐61029Italy
| | - Christian Celia
- Department of PharmacyUniversity of Chieti‐Pescara “G. D'Annunzio”Via dei Vestini 13ChietiI‐66100Italy
- Institute of Nanochemistry and NanobiologySchool of Environmental and Chemical EngineeringShanghai UniversityShanghai200444P. R. China
- Laboratory of Drug Targets HistopathologyInstitute of CardiologyLithuanian University of Health SciencesKaunasLT‐44307Lithuania
| | - Hélder A. Santos
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Department of Biomaterials and Biomedical TechnologyUniversity Medical Center Groningen, University of GroningenGroningen9713 AVThe Netherlands
| |
Collapse
|
3
|
Hussein HA, Khaphi FL. The Apoptotic Activity of Curcumin Against Oral Cancer Cells Without Affecting Normal Cells in Comparison to Paclitaxel Activity. Appl Biochem Biotechnol 2023; 195:5019-5033. [PMID: 37032374 DOI: 10.1007/s12010-023-04454-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 04/11/2023]
Abstract
Until now, chemotherapy, which has a series of side effects, has been the most widely employed treatment for different types of cancer. However, bioactive products have been utilized as alternative medicines for tumors due to their bioactivities with low or no side effects in normal cells. This research reported for the first time that curcumin (CUR) and paclitaxel (PTX) have significant anti-cancer activity against normal human gingival fibroblast (HGF) and tongue squamous cell carcinoma fibroblast (TSCCF) cell lines. The results showed that CUR (13.85 µg mL-1) and PTX (8.17 µg mL-1) significantly inhibited TSCCF cell viability, with no significant effect on normal HGF cells. SEM showed morphological changes in cells treated with CUR and PTX, especially with TSCCF cells, compared to HGF normal cells. For TSCCF, the results showed the highest necrosis was achieved with CUR (58.8%) and PTX (39%) as compared to the control (2.99%). For normal HGF cells, the highest early and late apoptosis was achieved with PTX. Further, DCFH-DA analyses showed no significant ROS stimulation in TSCCF and HGF cell lines treated with CUR and PTX. The 1H NMR analysis results show the presence of methoxy and hydroxyl groups and aromatic hydrogens in the CUR structure. In conclusion, the results confirmed that CUR is more specific to the oral cancer cells but not normal cells by inducing apoptosis in a dose- and time-dependent manner, with decreased TSCCF cell viability, and the cytotoxicity of CUR and PTX is not through the ROS pathway.
Collapse
Affiliation(s)
- Hanaa Ali Hussein
- College of Dentistry, University of Basrah 61004, Basic Science Branch, Al-Bara'iyah Street, Al-Sadir Teaching Hospital, Basrah city, 61001, Basrah, Iraq.
| | - Fatin L Khaphi
- College of Dentistry, University of Basrah 61004, Basic Science Branch, Al-Bara'iyah Street, Al-Sadir Teaching Hospital, Basrah city, 61001, Basrah, Iraq
| |
Collapse
|
4
|
Mishra AP, Singh P, Yadav S, Nigam M, Seidel V, Rodrigues CF. Role of the Dietary Phytochemical Curcumin in Targeting Cancer Cell Signalling Pathways. PLANTS (BASEL, SWITZERLAND) 2023; 12:plants12091782. [PMID: 37176840 PMCID: PMC10180989 DOI: 10.3390/plants12091782] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/19/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023]
Abstract
The diarylheptanoid curcumin [(1E,6E)-1,7-bis(4-hydroxy-3-methoxyphenyl)hepta-1,6-diene-3,5-dione] is one of the phenolic pigments responsible for the yellow colour of turmeric (Curcuma longa L.). This phytochemical has gained much attention in recent years due to its therapeutic potential in cancer. A range of drug delivery approaches have been developed to optimise the pharmacokinetic profile of curcumin and ensure that it reaches its target sites. Curcumin exhibits numerous biological effects, including anti-inflammatory, cardioprotective, antidiabetic, and anti-aging activities. It has also been extensively studied for its role as a cancer chemopreventive and anticancer agent. This review focusses on the role of curcumin in targeting the cell signalling pathways involved in cancer, particularly via modulation of growth factors, transcription factors, kinases and other enzymes, pro-inflammatory cytokines, and pro-apoptotic and anti-apoptotic proteins. It is hoped that this study will help future work on the potential of curcumin to fight cancer.
Collapse
Affiliation(s)
- Abhay Prakash Mishra
- Department of Pharmacology, Faculty of Health Science, University of Free State, Bloemfontein 9300, South Africa
| | - Pratichi Singh
- Department of Biosciences, School of Basic and Applied Sciences, Galgotias University, Greater Noida 203201, Uttar Pradesh, India
| | - Shikha Yadav
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, Uttar Pradesh, India
| | - Manisha Nigam
- Department of Biochemistry, H. N. B. Garhwal University, Srinagar Garhwal 246174, Uttarakhand, India
| | - Veronique Seidel
- Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Celia Fortuna Rodrigues
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- TOXRUN-Toxicology Research Unit, Cooperativa de Ensino Superior Politécnico e Universitário-CESPU, 4585-116 Gandra PRD, Portugal
| |
Collapse
|
5
|
Andreazzoli F, Bonucci M. Integrative Hematology: State of the Art. Int J Mol Sci 2023; 24:ijms24021732. [PMID: 36675247 PMCID: PMC9864076 DOI: 10.3390/ijms24021732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Blood cancers are a group of diseases with thus far frequently poor prognosis. Although many new drugs, including target therapies, have been developed in recent years, there is still a need to expand our therapeutic armamentarium to better deal with these diseases. Integrative hematology was conceived as a discipline that enriches the patient's therapeutic possibilities with the use of supplements, vitamins and a nutritional approach aiming at improving the response to therapies and the clinical outcome. We will analyze the substances that have proved most useful in preclinical and clinical studies in some of the most frequent blood diseases or in those where these studies are more numerous; the importance of the nutritional approach and the role of the intestinal microbiota will also be emphasized.
Collapse
Affiliation(s)
- Francesca Andreazzoli
- Department of Hematology, Versilia’s Hospital, Viale Aurelia, 335, 55049 Camaiore, Italy
- Correspondence:
| | - Massimo Bonucci
- Association for Research on Integrative Oncology Therapies (ARTOI), Via Ludovico Micara, 73, 00165 Rome, Italy
| |
Collapse
|
6
|
Azimirad M, Noori M, Azimirad F, Gholami F, Naseri K, Yadegar A, Asadzadeh Aghdaei H, Zali MR. Curcumin and capsaicin regulate apoptosis and alleviate intestinal inflammation induced by Clostridioides difficile in vitro. Ann Clin Microbiol Antimicrob 2022; 21:41. [PMID: 36155114 PMCID: PMC9511736 DOI: 10.1186/s12941-022-00533-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 09/01/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND The dramatic upsurge of Clostridioides difficile infection (CDI) by hypervirulent isolates along with the paucity of effective conventional treatment call for the development of new alternative medicines against CDI. The inhibitory effects of curcumin (CCM) and capsaicin (CAP) were investigated on the activity of toxigenic cell-free supernatants (Tox-S) of C. difficile RT 001, RT 126 and RT 084, and culture-filtrate of C. difficile ATCC 700057. METHODS Cell viability of HT-29 cells exposed to varying concentrations of CCM, CAP, C. difficile Tox-S and culture-filtrate was assessed by MTT assay. Anti-inflammatory and anti-apoptotic effects of CCM and CAP were examined by treatment of HT-29 cells with C. difficile Tox-S and culture-filtrate. Expression of BCL-2, SMAD3, NF-κB, TGF-β and TNF-α genes in stimulated HT-29 cells was measured using RT-qPCR. RESULTS C. difficile Tox-S significantly (P < 0.05) reduced the cell viability of HT-29 cells in comparison with untreated cells. Both CAP and CCM significantly (P < 0.05) downregulated the gene expression level of BCL-2, SMAD3, NF-κB and TNF-α in Tox-S treated HT-29 cells. Moreover, the gene expression of TGF-β decreased in Tox-S stimulated HT-29 cells by both CAP and CCM, although these reductions were not significantly different (P > 0.05). CONCLUSION The results of the present study highlighted that CCM and CAP can modulate the inflammatory response and apoptotic effects induced by Tox-S from different clinical C. difficile strains in vitro. Further studies are required to accurately explore the anti-toxin activity of natural components, and their probable adverse risks in clinical practice.
Collapse
Affiliation(s)
- Masoumeh Azimirad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Noori
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fahimeh Azimirad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Gholami
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kaveh Naseri
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
van Gils N, Verhagen HJ, Broux M, Martiáñez T, Denkers F, Vermue E, Rutten A, Csikós T, Demeyer S, Çil M, Al M, Cools J, Janssen JJ, Ossenkoppele GJ, Menezes RX, Smit L. Targeting histone methylation to reprogram the transcriptional state that drives survival of drug-tolerant myeloid leukemia persisters. iScience 2022; 25:105013. [PMID: 36097617 PMCID: PMC9463578 DOI: 10.1016/j.isci.2022.105013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/20/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Although chemotherapy induces complete remission in the majority of acute myeloid leukemia (AML) patients, many face a relapse. This relapse is caused by survival of chemotherapy-resistant leukemia (stem) cells (measurable residual disease; MRD). Here, we demonstrate that the anthracycline doxorubicin epigenetically reprograms leukemia cells by inducing histone 3 lysine 27 (H3K27) and H3K4 tri-methylation. Within a doxorubicin-sensitive leukemia cell population, we identified a subpopulation of reversible anthracycline-tolerant cells (ATCs) with leukemic stem cell (LSC) features lacking doxorubicin-induced H3K27me3 or H3K4me3 upregulation. These ATCs have a distinct transcriptional landscape than the leukemia bulk and could be eradicated by KDM6 inhibition. In primary AML, reprogramming the transcriptional state by targeting KDM6 reduced MRD load and survival of LSCs residing within MRD, and enhanced chemotherapy response in vivo. Our results reveal plasticity of anthracycline resistance in AML cells and highlight the potential of transcriptional reprogramming by epigenetic-based therapeutics to target chemotherapy-resistant AML cells. Reversible anthracycline-tolerant leukemia cells (ATCs) have low H3K27me3 or H3K4me3 ATCs exhibit stem cell features similar to leukemic stem cells Reprogramming the transcriptional state by inhibition of KDM6 depletes ATCs Inhibiting KDM6 adds to doxorubicin treatment and eradicates AML MRD (stem) cells
Collapse
|
8
|
Shaikh S, Shaikh J, Naba YS, Doke K, Ahmed K, Yusufi M. Curcumin: reclaiming the lost ground against cancer resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 4:298-320. [PMID: 35582033 PMCID: PMC9019276 DOI: 10.20517/cdr.2020.92] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/15/2020] [Accepted: 01/06/2021] [Indexed: 12/11/2022]
Abstract
Curcumin, a polyphenol, has a wide range of biological properties such as anticancer, antibacterial, antitubercular, cardioprotective and neuroprotective. Moreover, the anti-proliferative activities of Curcumin have been widely studied against several types of cancers due to its ability to target multiple pathways in cancer. Although Curcumin exhibited potent anticancer activity, its clinical use is limited due to its poor water solubility and faster metabolism. Hence, there is an immense interest among researchers to develop potent, water-soluble, and metabolically stable Curcumin analogs for cancer treatment. While drug resistance remains a major problem in cancer therapy that renders current chemotherapy ineffective, curcumin has shown promise to overcome the resistance and re-sensitize cancer to chemotherapeutic drugs in many studies. In the present review, we are summarizing the role of curcumin in controlling the proliferation of drug-resistant cancers and development of curcumin-based therapeutic applications from cell culture studies up to clinical trials.
Collapse
Affiliation(s)
- Siraj Shaikh
- Post-Graduate Department of Chemistry and Research Center, Abeda Inamdar Senior College of Arts, Science and Commerce (Affiliated to SPPU), Pune 411001, India.,Advanced Scientific Research Laboratory, Azam Campus, Pune 411001, India
| | - Javed Shaikh
- Post-Graduate Department of Chemistry and Research Center, Abeda Inamdar Senior College of Arts, Science and Commerce (Affiliated to SPPU), Pune 411001, India.,Advanced Scientific Research Laboratory, Azam Campus, Pune 411001, India
| | - Yusufi Sadia Naba
- Post-Graduate Department of Chemistry and Research Center, Abeda Inamdar Senior College of Arts, Science and Commerce (Affiliated to SPPU), Pune 411001, India
| | - Kailas Doke
- Post-Graduate Department of Chemistry and Research Center, Abeda Inamdar Senior College of Arts, Science and Commerce (Affiliated to SPPU), Pune 411001, India.,Advanced Scientific Research Laboratory, Azam Campus, Pune 411001, India
| | - Khursheed Ahmed
- Post-Graduate Department of Chemistry and Research Center, Abeda Inamdar Senior College of Arts, Science and Commerce (Affiliated to SPPU), Pune 411001, India.,Advanced Scientific Research Laboratory, Azam Campus, Pune 411001, India
| | - Mujahid Yusufi
- Post-Graduate Department of Chemistry and Research Center, Abeda Inamdar Senior College of Arts, Science and Commerce (Affiliated to SPPU), Pune 411001, India.,Advanced Scientific Research Laboratory, Azam Campus, Pune 411001, India
| |
Collapse
|
9
|
Vadukoot AK, Mottemmal S, Vekaria PH. Curcumin as a Potential Therapeutic Agent in Certain Cancer Types. Cureus 2022; 14:e22825. [PMID: 35399416 PMCID: PMC8980239 DOI: 10.7759/cureus.22825] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 12/17/2022] Open
Abstract
Cancer is a devastating disease condition and is the second most common etiology of death globally. After decades of research in the field of hematological malignancies and cellular therapeutics, we are still looking for therapeutic agents with the most efficacies and least toxicities. Curcumin is one of the cancer therapeutic agents that is derived from the Curcuma longa (turmeric) plant, and still in vitro and in vivo research is going on to find its beneficial effects on various cancers. Due to its potency to affect multiple targets of different cellular pathways, it is considered a promising agent to tackle various cancers alone or in combination with the existing chemotherapies. This review covers basic properties, mechanism of action, potential targets (molecules and cell-signaling pathways) of curcumin, as well as its effect on various solid and hematological malignancies.
Collapse
|
10
|
Yılmaz ZK, Özdemir Ö, Aslim B, Suludere Z, Şahin E. A new bio-active asymmetric-Schiff base: synthesis and evaluation of calf thymus DNA interaction, topoisomerase IIα inhibition, in vitro antiproliferative activity, SEM analysis and molecular docking studies. J Biomol Struct Dyn 2022; 41:2804-2822. [PMID: 35179080 DOI: 10.1080/07391102.2022.2039297] [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: 10/19/2022]
Abstract
In this paper, the asymmetric-Schiff base 2-(4-(2-hydroxybenzylideneamino)benzylideneamino)benzoic acid (SB-2) was newly synthesized and characterized by various spectroscopic methods. The interaction of SB-2 with calf thymus DNA was investigated by UV-vis, fluorescence spectroscopy and molecular docking methods. It was determined that SB-2 effectively binds to DNA via the intercalation mode. DNA electrophoretic mobility experiments displayed that topoisomerase IIα could not cleave pBR322 plasmid DNA in the presence of SB-2, confirming that the Schiff base acts as a topo II suppressor. In the molecular docking studies, SB-2 was found to show an affinity for both the DNA-topoisomerase IIα complex and the DNA. In vitro antiproliferative activity of SB-2 was screened against HT-29 (colorectal) and HeLa (cervical) human tumor cell lines by MTT assay. SB-2 diminished the cell viability in a concentration- and incubation time-dependent manner. The ability of SB-2 to measure DNA damage in tumor cells was evaluated with cytokinesis-block micronucleus assay after incubation 24 h and 48 h. Light and scanning electron microscopy experiments of tumor cells demonstrated an incubation time-dependent increase in the proportion of apoptotic cells (nuclear condensation and apoptotic bodies) suggesting that autophagy and apoptosis play a role in the death of cells. Based on the obtained results, it may be considered that SB-2 is a candidate for DNA-targeting antitumor drug.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Zehra Kübra Yılmaz
- Department of Biology, Faculty of Science, Gazi University, Ankara, Turkey
| | - Özlem Özdemir
- Department of Chemistry, Faculty of Science, Gazi University, Ankara, Turkey
| | - Belma Aslim
- Department of Biology, Faculty of Science, Gazi University, Ankara, Turkey
| | - Zekiye Suludere
- Department of Biology, Faculty of Science, Gazi University, Ankara, Turkey
| | - Egemen Şahin
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
11
|
Park N, Park Y, Yadav AK, Shin Y, Bishop‐Bailey D, Choi J, Park JW, Jang B. Anti-growth and pro-apoptotic effects of dasatinib on human oral cancer cells through multi-targeted mechanisms. J Cell Mol Med 2021; 25:8300-8311. [PMID: 34318593 PMCID: PMC8419177 DOI: 10.1111/jcmm.16782] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/29/2021] [Accepted: 06/15/2021] [Indexed: 12/17/2022] Open
Abstract
Dasatinib is an inhibitor of Src that has anti-tumour effects on many haematological and solid cancers. However, the anti-tumour effects of dasatinib on human oral cancers remain unclear. In this study, we investigated the effects of dasatinib on different types of human oral cancer cells: the non-tumorigenic YD-8 and YD-38 and the tumorigenic YD-10B and HSC-3 cells. Strikingly, dasatinib at 10 µM strongly suppressed the growth and induced apoptosis of YD-38 cells and inhibited the phosphorylation of Src, EGFR, STAT-3, STAT-5, PKB and ERK-1/2. In contrast, knockdown of Src blocked the phosphorylation of EGFR, STAT-5, PKB and ERK-1/2, but not STAT-3, in YD-38 cells. Dasatinib induced activation of the intrinsic caspase pathway, which was inhibited by z-VAD-fmk, a pan-caspase inhibitor. Dasatinib also decreased Mcl-1 expression and S6 phosphorylation while increased GRP78 expression and eIF-2α phosphorylation in YD-38 cells. In addition, to its direct effects on YD-38 cells, dasatinib also exhibited anti-angiogenic properties. Dasatinib-treated YD-38 or HUVEC showed reduced HIF-1α expression and stability. Dasatinib alone or conditioned media from dasatinib-treated YD-38 cells inhibited HUVEC tube formation on Matrigel without affecting HUVEC viability. Importantly, dasatinib's anti-growth, anti-angiogenic and pro-apoptotic effects were additionally seen in tumorigenic HSC-3 cells. Together, these results demonstrate that dasatinib has strong anti-growth, anti-angiogenic and pro-apoptotic effects on human oral cancer cells, which are mediated through the regulation of multiple targets, including Src, EGFR, STAT-3, STAT-5, PKB, ERK-1/2, S6, eIF-2α, GRP78, caspase-9/3, Mcl-1 and HIF-1α.
Collapse
Affiliation(s)
- Nam‐Sook Park
- Department of Molecular MedicineCollege of MedicineKeimyung UniversityDaeguKorea
| | - Yu‐Kyung Park
- Department of Molecular MedicineCollege of MedicineKeimyung UniversityDaeguKorea
| | - Anil Kumar Yadav
- Department of Molecular MedicineCollege of MedicineKeimyung UniversityDaeguKorea
| | - Young‐Min Shin
- Department of DentistryCollege of MedicineKeimyung UniversityDaeguKorea
| | | | - Jong‐Soon Choi
- Biological Disaster Analysis GroupDivision of Convergence BiotechnologyKorea Basic Science InstituteDaejeonKorea
- Graduate School of Analytical Science and TechnologyChungnam National UniversityDaejeonKorea
| | - Jong Wook Park
- Department of ImmunologyCollege of MedicineKeimyung UniversityDaeguKorea
| | - Byeong‐Churl Jang
- Department of Molecular MedicineCollege of MedicineKeimyung UniversityDaeguKorea
| |
Collapse
|
12
|
Zoi V, Galani V, Lianos GD, Voulgaris S, Kyritsis AP, Alexiou GA. The Role of Curcumin in Cancer Treatment. Biomedicines 2021; 9:1086. [PMID: 34572272 PMCID: PMC8464730 DOI: 10.3390/biomedicines9091086] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022] Open
Abstract
Curcumin is a polyphenol extracted from the rhizomes of the turmeric plant, Curcuma longa which has anti-inflammatory, and anticancer properties. Chronic inflammation is associated with the development of cancer. Curcumin acts on the regulation of various immune modulators, including cytokines, cyclooxygenase-2 (COX-2), and reactive oxygen species (ROS), which partly explains its anticancer effects. It also takes part in the downregulation of growth factors, protein kinases, oncogenic molecules and various signaling pathways, such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), c-Jun N-terminal kinase (JNK) and signal transducer and activator of transcription 3 (STAT3) signaling. Clinical trials of curcumin have been completed or are ongoing for various types of cancer. This review presents the molecular mechanisms of curcumin in different types of cancer and the evidence from the most recent clinical trials.
Collapse
Affiliation(s)
- Vasiliki Zoi
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
- Department of Anatomy Histology-Embryology, School of Medicine, University of Ioannina, 45500 Ioannina, Greece;
| | - Vasiliki Galani
- Department of Anatomy Histology-Embryology, School of Medicine, University of Ioannina, 45500 Ioannina, Greece;
| | - Georgios D. Lianos
- Department of Surgery, University Hospital of Ioannina, 45500 Ioannina, Greece;
| | - Spyridon Voulgaris
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
- Department of Neurosurgery, School of Medicine Ioannina, University of Ioannina, 45500 Ioannina, Greece
| | - Athanasios P. Kyritsis
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
| | - George A. Alexiou
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
- Department of Neurosurgery, School of Medicine Ioannina, University of Ioannina, 45500 Ioannina, Greece
| |
Collapse
|
13
|
Shetty NP, Prabhakaran M, Srivastava AK. Pleiotropic nature of curcumin in targeting multiple apoptotic-mediated factors and related strategies to treat gastric cancer: A review. Phytother Res 2021; 35:5397-5416. [PMID: 34028111 DOI: 10.1002/ptr.7158] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/21/2021] [Accepted: 04/30/2021] [Indexed: 12/24/2022]
Abstract
Gastric cancer (GC) is one of the major reasons for cancer-associated death and exhibits the second-highest mortality rate worldwide. Several advanced approaches have been designed to treat GC; however, these strategies possess many innate complications. In view of this, the upcoming research relying on natural products could result in designing potential anticancer agents with fewer side effects. Curcumin, isolated from the rhizomes of Curcuma longa L. has several medicinal properties like antiinflammatory, antioxidant, antiapoptotic, antitumor, and antimetastatic. Such pleiotropic nature of curcumin impedes the invasion and proliferation of GC by targeting several oncogenic factors like p23, human epidermal factor receptor2 including Helicobacter pylori. The side effect of chemotherapy, that is, chemotherapeutic resistance and radiotherapy could be reduced combination therapy of curcumin. Moreover, the photodynamic therapy of curcumin destroys the cancer cells without affecting normal cells. However, further more potential studies are required to establish the potent efficacy of curcumin in the treatment of GC. The current review details the anticancer activities of curcumin and related strategies which could be employed to treat GC with additional focus on its inhibitory properties against viability, proliferation, and migration of GC cells through cell cycle arrest and stimulation by apoptosis-mediated factors.
Collapse
Affiliation(s)
- Nandini P Shetty
- Plant Cell Biotechnology Department, CSIR-Central Food Technological Research Institute, Mysore, 570020, India
| | - Manoj Prabhakaran
- Plant Cell Biotechnology Department, CSIR-Central Food Technological Research Institute, Mysore, 570020, India
| | | |
Collapse
|
14
|
Al-Aamri HM, Irving HR, Bradley C, Meehan-Andrews T. Intrinsic and extrinsic apoptosis responses in leukaemia cells following daunorubicin treatment. BMC Cancer 2021; 21:438. [PMID: 33879127 PMCID: PMC8059319 DOI: 10.1186/s12885-021-08167-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 04/06/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Daunorubicin is used clinically in the treatment of myeloma, acute lymphatic and myelocytic leukaemia. The toxic lesions caused by daunorubicin induce various modes of cell death, including apoptosis. Apoptosis is highly regulated programmed cell death that can be initiated mainly via two pathways, through death receptors (extrinsic) or involvement of the mitochondria (intrinsic). Induction of apoptosis via these pathways has been alluded following treatment with daunorubicin, but never compared in acute lymphoblastic leukaemia over a time course. METHODS This study investigated the mechanisms of daunorubicin induced apoptosis in the treatment of CCRF-CEM, MOLT-4 (acute T-lymphoblastic leukaemia) and SUP-B15 (acute B-lymphoblastic leukaemia) cells. Cells were treated with daunorubicin for 4 h, and then placed in recovery medium (without daunorubicin) for 4 h, 12 h and 24 h. Apoptotic response was analysing using annexin-V expression, caspase activity, mitochondrial membrane potential change and an array to detect 43 apoptotic proteins. RESULTS Daunorubicin induced apoptosis in all leukemic cell lines, but with different levels and duration of response. Both apoptosis levels and caspase activity increased after four hours recovery then declined in CCRF-CEM and MOLT-4 cells. However, SUP-B15 cells displayed initially comparable levels but remained elevated over the 24 h assessment period. Changes in mitochondrial membrane potential occurred in both MOLT-4 and CCRF-CEM cells but not in SUP-B15 cells. Expression of apoptotic proteins, including Bcl-2, Bax, caspase 3 and FADD, indicated that daunorubicin potentially induced both extrinsic and intrinsic apoptosis in both CCRF-CEM and MOLT-4 cells, but only extrinsic apoptosis in SUP-B15 cells. CONCLUSIONS This study describes variations in sensitivities and timing of apoptotic responses in different leukaemia cell lines. These differences could be attributed to the lack of functional p53 in coordinating the cells response following cytotoxic treatment with daunorubicin, which appears to delay apoptosis and utilises alternative signalling mechanisms that need to be further explored.
Collapse
Affiliation(s)
- Hussain Mubarak Al-Aamri
- Department of Pharmacy and Biomedical Science, La Trobe Institute for Molecular Science (LIMS), La Trobe University, P.O. Box 199, Bendigo, VIC, 3552, Australia.,Oman College of Health Sciences, PO Box 293, 620, Ruwi, Sultanate of Oman
| | - Helen R Irving
- Department of Pharmacy and Biomedical Science, La Trobe Institute for Molecular Science (LIMS), La Trobe University, P.O. Box 199, Bendigo, VIC, 3552, Australia
| | - Christopher Bradley
- Department of Pharmacy and Biomedical Science, La Trobe Institute for Molecular Science (LIMS), La Trobe University, P.O. Box 199, Bendigo, VIC, 3552, Australia
| | - Terri Meehan-Andrews
- Department of Pharmacy and Biomedical Science, La Trobe Institute for Molecular Science (LIMS), La Trobe University, P.O. Box 199, Bendigo, VIC, 3552, Australia.
| |
Collapse
|
15
|
Design, Synthesis and Evaluation of New Bioactive Oxadiazole Derivatives as Anticancer Agents Targeting Bcl-2. Int J Mol Sci 2020; 21:ijms21238980. [PMID: 33256166 PMCID: PMC7730549 DOI: 10.3390/ijms21238980] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 01/05/2023] Open
Abstract
A series of 2-(1H-indol-3-yl)-5-substituted-1,3,4-oxadiazoles, 4a-m, were designed, synthesized and tested in vitro as potential pro-apoptotic Bcl-2 inhibitory anticancer agents based on our previously reported hit compounds. Synthesis of the target 1,3,4-oxadiazoles was readily accomplished through a cyclization reaction of indole carboxylic acid hydrazide 2 with substituted carboxylic acid derivatives 3a-m in the presence of phosphorus oxychloride. New compounds 4a-m showed a range of IC50 values concentrated in the low micromolar range selectively in Bcl-2 positive human cancer cell lines. The most potent candidate 4-trifluoromethyl substituted analogue 4j showed selective IC50 values of 0.52-0.88 μM against Bcl-2 expressing cell lines with no inhibitory effects in the Bcl-2 negative cell line. Moreover, 4j showed binding that was two-fold more potent than the positive control gossypol in the Bcl-2 ELISA binding affinity assay. Molecular modeling studies helped to further rationalize anti-apoptotic Bcl-2 binding and identified compound 4j as a candidate with drug-like properties for further investigation as a selective Bcl-2 inhibitory anticancer agent.
Collapse
|
16
|
Chen Y, Zheng J, Gan D, Chen Y, Zhang N, Chen Y, Lin Z, Wang W, Chen H, Lin D, Hu J. E35 ablates acute leukemia stem and progenitor cells in vitro and in vivo. J Cell Physiol 2020; 235:8023-8034. [PMID: 31960417 PMCID: PMC7540425 DOI: 10.1002/jcp.29457] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 01/03/2020] [Indexed: 12/11/2022]
Abstract
Leukemia stem cells (LSCs) have critical functions in acute leukemia (AL) pathogenesis, participating in its initiation and relapse. Thus, identifying new molecules to eradicate LSCs represents a high priority for AL management. This work identified E35, a novel Emodin derivative, which strongly inhibited growth and enhanced apoptosis of AL stem cell lines, and primary stem and progenitor cells from AL cases, while sparing normal hematopoietic cells. Furthermore, functional assays in cultured cells and animals suggested that E35 preferentially ablated primitive leukemia cell populations without impairing their normal counterparts. Moreover, molecular studies showed that E35 remarkably downregulated drug-resistant gene and dramatically inhibited the Akt/mammalian target of rapamycin signaling pathway. Notably, the in vivo anti-LSC activity of E35 was further confirmed in murine xenotransplantation models. Collectively, these findings indicate E35 constitutes a novel therapeutic candidate for AL, potentially targeting leukemia stem and progenitor cells.
Collapse
Affiliation(s)
- Yingyu Chen
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
| | - Jing Zheng
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
| | - Donghui Gan
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
- Department of HematologyThe Affiliated Hospital of Putian UniversityPutianFujianChina
| | - Yanxin Chen
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
| | - Na Zhang
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
| | - Yuwen Chen
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
| | - Zhenxing Lin
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
| | - Wenfeng Wang
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of ChemistryFuzhou UniversityFuzhouFujianChina
| | - Haijun Chen
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of ChemistryFuzhou UniversityFuzhouFujianChina
| | - Donghong Lin
- Department of Clinical LaboratorySchool of Medical Technology and EngineeringFujian Medical UniversityFujianChina
| | - Jianda Hu
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
| |
Collapse
|
17
|
Hafizi M, Kalanaky S, Fakharzadeh S, Janzamin E, Arjmandi T, Atashi A, Nazaran MH. GFc7 as a Smart Growth Nanofactor for ex vivo Expansion and Cryoprotection of Humans' Hematopoietic Stem Cells. Int J Nanomedicine 2020; 15:6263-6277. [PMID: 32922002 PMCID: PMC7457843 DOI: 10.2147/ijn.s256104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
Background Nowadays, smart synthesized nanostructures have attracted wide attention in the field of stem cell nanotechnology due to their effect on different properties of stem cells. Methods GFc7 growth nanofactor was synthesized based on nanochelating technology as an iron-containing copper chelator nanocomplex. The effect of this nanocomplex on the expansion and differentiation of hematopoietic stem cells (HSCs) as well as its performance as a cryoprotectant was evaluated in the present study. Results The results showed that the absolute count of CD34+ and CD34+CD38- cells on days 4, 7, 10 and 13; the percentage of lactate dehydrogenase enzyme on the same days and CD34+CXCR4 population on day 10 were significantly increased when they were treated with GFc7 growth nanofactor in a fetal bovine serum (FBS)-free medium. This medium also led to delayed differentiation in HSCs. One noticeable result was that CD34+CD38- cells cultured in an FBS medium were immediately differentiated into CD34+CD38+ cells, while CD34+CD38- cells treated with GFc7 growth nanofactor in FBS medium did not show such an immediate significant differentiation. De-freezing GFc7-treated CD34+ cells, which were already frozen according to cord blood bank protocols, showed a higher percentage of cell viability and a larger number of colonies according to colony-forming cell assay as compared to control. Conclusion It can be claimed that treating HSCs with GFc7 growth nanofactor leads to quality and quantity improvement of HSCs, both in terms of expansion in vitro and freezing and de-freezing processes.
Collapse
Affiliation(s)
- Maryam Hafizi
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Somayeh Kalanaky
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Saideh Fakharzadeh
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | | | - Tarlan Arjmandi
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Amir Atashi
- Stem Cell and Tissue Engineering Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | | |
Collapse
|
18
|
Physicochemical characterization of polysaccharide from the leaf of Dendrobium officinale and effect on LPS induced damage in GES-1 cell. Int J Biol Macromol 2020; 149:320-330. [DOI: 10.1016/j.ijbiomac.2020.01.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/25/2019] [Accepted: 01/04/2020] [Indexed: 01/08/2023]
|
19
|
Ahmed N, Kermanshahi B, Ghazani SM, Tait K, Tcheng M, Roma A, Callender SP, Smith RW, Tam W, Wettig SD, Rogers MA, Marangoni AG, Spagnuolo PA. Avocado-derived polyols for use as novel co-surfactants in low energy self-emulsifying microemulsions. Sci Rep 2020; 10:5566. [PMID: 32221368 PMCID: PMC7101315 DOI: 10.1038/s41598-020-62334-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 03/09/2020] [Indexed: 12/20/2022] Open
Abstract
Avocado (Persea americana Mill.; Lauraceae) seed-derived polyhydroxylated fatty alcohols (PFAs) or polyols (i.e., avocadene and avocadyne) are metabolic modulators that selectively induce apoptosis of leukemia stem cells and reverse pathologies associated with diet-induced obesity. Delivery systems containing avocado polyols have not been described. Herein, natural surface active properties of these polyols are characterized and incorporated into self-emulsifying drug delivery systems (SEDDS) that rely on molecular self-assembly to form fine, transparent, oil-in-water (O/W) microemulsions as small as 20 nanometers in diameter. Mechanistically, a 1:1 molar ratio of avocadene and avocadyne (i.e., avocatin B or AVO was shown to be a eutectic mixture which can be employed as a novel, bioactive, co-surfactant that significantly reduces droplet size of medium-chain triglyceride O/W emulsions stabilized with polysorbate 80. In vitro cytotoxicity of avocado polyol-SEDDS in acute myeloid leukemia cell lines indicated significant increases in potency and bioactivity compared to conventional cell culture delivery systems. A pilot pharmacokinetic evaluation of AVO SEDDS in C57BL/6J mice revealed appreciable accumulation in whole blood and biodistribution in key target tissues. Lastly, incorporation of AVO in SEDDS significantly improved encapsulation of the poorly water-soluble drugs naproxen and curcumin.
Collapse
Affiliation(s)
- Nawaz Ahmed
- Department of Food Science, University of Guelph, Guelph, Ontario, N1G 2WI, Canada
| | - Behnoush Kermanshahi
- Department of Food Science, University of Guelph, Guelph, Ontario, N1G 2WI, Canada
| | - Saeed M Ghazani
- Department of Food Science, University of Guelph, Guelph, Ontario, N1G 2WI, Canada
| | - Katrina Tait
- Department of Food Science, University of Guelph, Guelph, Ontario, N1G 2WI, Canada
| | - Matthew Tcheng
- Department of Food Science, University of Guelph, Guelph, Ontario, N1G 2WI, Canada
| | - Alessia Roma
- Department of Food Science, University of Guelph, Guelph, Ontario, N1G 2WI, Canada
| | - Shannon P Callender
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| | - Richard W Smith
- University of Waterloo Mass Spectrometry Facility, Department of Chemistry, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - William Tam
- Guelph-Waterloo Centre for Graduate Work in Chemistry and Biochemistry, Department of Chemistry, University of Guelph, Guelph, Ontario, N1G 2W1, Canada
| | - Shawn D Wettig
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
- Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Ave. W., Waterloo, Ontario, N2L 3G1, Canada
| | - Michael A Rogers
- Department of Food Science, University of Guelph, Guelph, Ontario, N1G 2WI, Canada
| | | | - Paul A Spagnuolo
- Department of Food Science, University of Guelph, Guelph, Ontario, N1G 2WI, Canada.
| |
Collapse
|
20
|
Samarkhazan NS, Yekta R, Sayadi M, Tackallou SH, Safaralizadeh R, Mahdavi M. 2-NDC from dithiocarbamates improves ATRA efficiency and ROS-induced apoptosis via downregulation of Bcl2 and Survivin in human acute promyelocytic NB4 cells. Hum Exp Toxicol 2020; 39:960-972. [PMID: 32096428 DOI: 10.1177/0960327120905958] [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] [Indexed: 11/15/2022]
Abstract
Although it has been widely considered that all-trans retinoic acid (ATRA) is an efficient therapeutic agent for acute promyelocytic leukemia (APL), there is an urgent need for extending and examining new therapeutics in medicine. Dithiocarbamates (DTCs) are one of the recent important chemical synthetic compounds used in cancer therapy. The aim of this study was to evaluate the apoptosis-inducing effect of 2-nitro-1-phenylethylpiperidine-1-carbodithioate (2-NDC) as an active derivative from DTCs, in combination with ATRA on human APL NB4 cells. The viability of treated NB4 cells was measured by 3-(4,5-dimethyltiazol-2-yl)-2,5-diphenyltetrazolium bromide assay in various concentrations (10-120 µM). The proapoptotic effects of 2-NDC were investigated by acridine orange/ethidium bromide staining, DNA ladder formation, and flow cytometry. We also assessed the oxidative stress-inducing effect of 2-NDC and in combination with ATRA on the NB4 cells. The alteration in gene expression levels of Bax, Bcl2, and Survivin was measured through a real-time polymerase chain reaction. Furthermore, we redetected the interaction between 2-NDC and antiapoptotic proteins Bcl2 and Survivin via molecular docking. We found that 2-NDC induced apoptosis in NB4 cells in a time-dosage-dependent manner. Also, 2-NDC triggered apoptosis by expanding intracellular reactive oxygen species, combined with ATRA. Bax/Bcl2 ratio was modulated and Survivin was downregulated in NB4 cells upon 2-NDC treatment. Molecular docking studies indicated that 2-NDC binds to the baculovirus inhibitor of apoptosis protein repeat domain of Survivin and Bcl homology 3 domain of Bcl2 with various affinities. Based on the present observations, it seems that this derivative can be estimated as an appropriate candidate for future pharmaceutical evaluations.
Collapse
Affiliation(s)
- N S Samarkhazan
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - R Yekta
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - M Sayadi
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - S H Tackallou
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - R Safaralizadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - M Mahdavi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
21
|
Mohammadi Kian M, Salemi M, Bahadoran M, Haghi A, Dashti N, Mohammadi S, Rostami S, Chahardouli B, Babakhani D, Nikbakht M. Curcumin Combined with Thalidomide Reduces Expression of STAT3 and Bcl-xL, Leading to Apoptosis in Acute Myeloid Leukemia Cell Lines. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:185-194. [PMID: 32021103 PMCID: PMC6970263 DOI: 10.2147/dddt.s228610] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 12/16/2019] [Indexed: 12/29/2022]
Abstract
Introduction Acute myeloid leukemia (AML) is a type of blood disorder that exhibits uncontrolled growth and reduced ability to undergo apoptosis. Signal transducer and activator of transcription 3 (STAT3) is a family member of transcription factors which promotes carcinogenesis in most human cancers. This effect on AML is accomplished through deregulation of several critical genes, such as B cell lymphoma-extra-large (BCL-XL) which is anti-apoptotic protein. The aim of this study was to evaluate the effect of curcumin (CUR) and thalidomide (THAL) on apoptosis induction and also the alteration of the mRNA expression level of STAT3 and BCL-XL mRNA on AML cell line compounds. Methods The growth inhibitory effects of CUR and THAL and their combination were measured by MTT assay in U937 and KG-1 cell lines. The rates of apoptosis induction and cell cycle analysis were measured by concurrent staining with Annexin V and PI. The mRNA expression level of STAT3 and BCL-XL was evaluated by Real-Time PCR. Results CUR inhibited proliferation and induced apoptosis in both KG-1 and U937 cells and this effect increased by combination with THAL. The expression level of STAT3 and BCL-XL was significantly down-regulated in KG-1 cells after treatment by CUR and THAL and their combination. Conclusion Overall, our findings suggested that down-regulation of STAT3 and BCL-XL mRNA expression in response to CUR and THAL treatment lead to inhibition of cell growth and induction of apoptosis.
Collapse
Affiliation(s)
- Mahnaz Mohammadi Kian
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdieh Salemi
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Bahadoran
- Department of Biochemistry, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Atousa Haghi
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Young Researchers & Elite Club Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nasrin Dashti
- Department of Medical Laboratory Sciences, School of Allied Health Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Mohammadi
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahrbano Rostami
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahram Chahardouli
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Babakhani
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Nikbakht
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Cho H, Yadav AK, Do Y, Heo M, Bishop-Bailey D, Lee J, Jang BC. Anti‑survival and pro‑apoptotic effects of meridianin C derivatives on MV4‑11 human acute myeloid leukemia cells. Int J Oncol 2019; 56:368-378. [PMID: 31789392 DOI: 10.3892/ijo.2019.4925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 10/24/2019] [Indexed: 11/06/2022] Open
Abstract
Meridianin C is a marine natural product with anticancer activity. Several meridianin C derivatives (compounds 7a‑j) were recently synthesized, and their inhibitory effects on pro‑viral integration site for Moloney murine leukemia virus (PIM) kinases, as well as their antiproliferative effects on human leukemia cells, were reported. However, the anti‑leukemic effects and mechanisms of action of meridianin C and its derivatives remain largely unknown. The aim of the present study was to investigate the effects of meridianin C and its derivatives on MV4‑11 human acute myeloid leukemia cell growth. The parent compound meridianin C did not markedly affect the viability and survival of MV4‑11 cells. By contrast, MV4‑11 cell viability and survival were reduced by meridianin C derivatives, with compound 7a achieving the most prominent reduction. Compound 7a notably inhibited the expression and activity of PIM kinases, as evidenced by reduced B‑cell lymphoma‑2 (Bcl‑2)‑associated death promoter phosphorylation at Ser112. However, meridianin C also suppressed PIM kinase expression and activity, and the pan‑PIM kinase inhibitor AZD1208 only slightly suppressed the survival of MV4‑11 cells. Thus, the anti‑survival effect of compound 7a on MV4‑11 cells was unrelated to PIM kinase inhibition. Moreover, compound 7a induced apoptosis, caspase‑9 and ‑3 activation and poly(ADP‑ribose) polymerase (PARP) cleavage, but did not affect death receptor (DR)‑4 or DR‑5 expression in MV4‑11 cells. Compound 7a also induced the generation of cleaved Bcl‑2, and the downregulation of myeloid cell leukemia (Mcl)‑1 and X‑linked inhibitor of apoptosis (XIAP) in MV4‑11 cells. Furthermore, compound 7a increased eukaryotic initiation factor (eIF)‑2α phosphorylation and decreased S6 phosphorylation, whereas GRP‑78 expression was unaffected. Importantly, treatment with a pan‑caspase inhibitor (z‑VAD‑fmk) significantly attenuated compound 7a‑induced apoptosis, caspase‑9 and ‑3 activation, PARP cleavage, generation of cleaved Bcl‑2 and downregulation of Mcl‑1 and XIAP in MV4‑11 cells. Collectively, these findings demonstrated the strong anti‑survival and pro‑apoptotic effects of compound 7a on MV4‑11 cells through regulation of caspase‑9 and ‑3, Bcl‑2, Mcl‑1, XIAP, eIF‑2α and S6 molecules.
Collapse
Affiliation(s)
- Hyorim Cho
- Department of Molecular Medicine, College of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| | - Anil Kumar Yadav
- Department of Molecular Medicine, College of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| | - Youngrok Do
- Department of Hematology and Oncology, College of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| | - Mihwa Heo
- Department of Hematology and Oncology, College of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| | - David Bishop-Bailey
- Comparative Biomedical Sciences, Royal Veterinary College, London NW 10TU, United Kingdom
| | - Jinho Lee
- Department of Chemistry, College of Life Science, Keimyung University, Daegu 42601, Republic of Korea
| | - Byeong-Churl Jang
- Department of Molecular Medicine, College of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| |
Collapse
|
23
|
Ledesma-Martínez E, Aguíñiga-Sánchez I, Weiss-Steider B, Rivera-Martínez AR, Santiago-Osorio E. Casein and Peptides Derived from Casein as Antileukaemic Agents. JOURNAL OF ONCOLOGY 2019; 2019:8150967. [PMID: 31582978 PMCID: PMC6754885 DOI: 10.1155/2019/8150967] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/20/2019] [Accepted: 08/04/2019] [Indexed: 12/20/2022]
Abstract
Milk is a heterogeneous lacteal secretion mixture of numerous components that exhibit a wide variety of chemical and functional activities. Casein, the main protein in milk, is composed of α-, β-, and κ-caseins, each of which is important for nutritional value and for promoting the release of cytokines, also are linked to the regulation of haematopoiesis and immune response and inhibit the proliferation and induce the differentiation of leukaemia cells. It has been shown that the digestive process of caseins leads to the release of bioactive peptides that are involved in the regulation of blood pressure and the inhibition or activation of the immune response by serving as agonists or antagonists of opioid receptors, thus controlling the expression of genes that exert epigenetic control. Later, they bind to opioid receptor, block nuclear factor κ-beta, increase the redox potential, and reduce oxidative stress and the pro-inflammatory agents that favour an antioxidant and anti-inflammatory environment. Therefore, the bioactive peptides of casein could be compounds with antileukaemia potential. This review provides a summary of current knowledge about caseins and casein peptides on the immune system as well as their roles in the natural defence against the development of leukaemia and as relevant epigenetic regulators that can help eradicate leukaemia.
Collapse
Affiliation(s)
- Edgar Ledesma-Martínez
- Haematopoiesis and Leukaemia Laboratory, Research Unit on Cell Differentiation and Cancer, FES Zaragoza, National Autonomous University of Mexico, 09230 Mexico City, Mexico
| | - Itzen Aguíñiga-Sánchez
- Haematopoiesis and Leukaemia Laboratory, Research Unit on Cell Differentiation and Cancer, FES Zaragoza, National Autonomous University of Mexico, 09230 Mexico City, Mexico
| | - Benny Weiss-Steider
- Haematopoiesis and Leukaemia Laboratory, Research Unit on Cell Differentiation and Cancer, FES Zaragoza, National Autonomous University of Mexico, 09230 Mexico City, Mexico
| | - Ana Rocío Rivera-Martínez
- Haematopoiesis and Leukaemia Laboratory, Research Unit on Cell Differentiation and Cancer, FES Zaragoza, National Autonomous University of Mexico, 09230 Mexico City, Mexico
| | - Edelmiro Santiago-Osorio
- Haematopoiesis and Leukaemia Laboratory, Research Unit on Cell Differentiation and Cancer, FES Zaragoza, National Autonomous University of Mexico, 09230 Mexico City, Mexico
| |
Collapse
|
24
|
New Quinoline-Based Heterocycles as Anticancer Agents Targeting Bcl-2. Molecules 2019; 24:molecules24071274. [PMID: 30986908 PMCID: PMC6479519 DOI: 10.3390/molecules24071274] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/21/2019] [Accepted: 03/28/2019] [Indexed: 11/18/2022] Open
Abstract
The Bcl-2 protein has been studied as an anticancer drug target in recent years, due to its gatekeeper role in resisting programmed cancer cell death (apoptosis), and the design of BH3 domain mimetics has led to the clinical approval of Venetoclax (ABT-199) for the treatment of chronic lymphocytic leukaemia. In this work we extend our previous studies on the discovery of indole-based heterocycles as Bcl-2 inhibitors, to the identification of quinolin-4-yl based oxadiazole and triazole analogues. Target compounds were readily synthesized via a common aryl-substituted quinolin-4-carbonyl-N-arylhydrazine-1-carbothioamide (5a–b) intermediate, through simple variation of the basic cyclisation conditions. Some of the quinoline-based oxadiazole analogues (e.g. compound 6i) were found to exhibit sub-micromolar anti-proliferative activity in Bcl-2-expressing cancer cell lines, and sub-micromolar IC50 activity within a Bcl2-Bim peptide ELISA assay. The Bcl-2 targeted anticancer activity of 6i was further rationalised via computational molecular modelling, offering possibilities to extend this work into the design of further potent and selective Bcl-2 inhibitory heteroaromatics with therapeutic potential.
Collapse
|
25
|
Zahedpanah M, Takanlu JS, Nikbakht M, Rad F, Farhid F, Mousavi SA, Rad S, Fumani HK, Hosseini Rad SMA, Mohammadi S. Microvesicles of osteoblasts modulate bone marrow mesenchymal stem cell-induced apoptosis to curcumin in myeloid leukemia cells. J Cell Physiol 2019; 234:18707-18719. [PMID: 30916405 DOI: 10.1002/jcp.28511] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 02/16/2019] [Accepted: 02/20/2019] [Indexed: 11/07/2022]
Abstract
Microvesicles (MVs) derived from bone marrow niche components have an important role in genetic reprogramming and subsequent drugs induce apoptosis in leukemic cells. Here, we have found that undertreatment of curcumin or daunorubicin, the cross-talk through MVs of KG-1-bone marrow mesenchymal stem cells (BMSCs), significantly downregulates the expression of the survival gene osteopontin (OPN), CXCL-12, IL-6 (interleukin-6), STAT-3, and VCAM-1 (vascular cell adhesion molecule 1) in treated-KG-1 cells as well as exclusively upregulates CXCL-12 in BMSCs. Drug treated-cell populations' MVs of both single cultured osteoblasts (OBs) and cocultured KG-1 + BMSCs + OBs similarly upregulate survival mediators' OPN, CXCL-12, IL-6, STAT-3, and VCAM-1 in treated-KG-1 cells. Likewise, isolated MVs from KG-1 cells or communication between KG-1, BMSCs, and OBs treated by drugs increase the expression of genes OPN, CXCL-12, IL-6, STAT3, and VCAM-1 by OBs. MVs derived from KG-1 + BMSCs + OBs reduce drug-induced apoptosis in KG-1 cells. This suggests MVs-mediated information transfer is a procedure whereby OBs could overcome BMSCs-induced apoptosis in drug-treated-KG-1 cells.
Collapse
Affiliation(s)
- Mahdi Zahedpanah
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Javid Sabour Takanlu
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| | - Mohsen Nikbakht
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| | - Fariba Rad
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Fatemeh Farhid
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Asadollah Mousavi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| | - Soroush Rad
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| | - Hosein Kamranzadeh Fumani
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| | | | - Saeed Mohammadi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| |
Collapse
|
26
|
Kouhpeikar H, Butler AE, Bamian F, Barreto GE, Majeed M, Sahebkar A. Curcumin as a therapeutic agent in leukemia. J Cell Physiol 2019; 234:12404-12414. [PMID: 30609023 DOI: 10.1002/jcp.28072] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 12/18/2018] [Indexed: 12/28/2022]
Abstract
Leukemia comprises a group of hematological malignancies responsible for 8% of all cancers and is the most common cancer in children. Despite significant improvements in leukemia treatment, the efficacy of conventional chemotherapeutic agents is low and the disease carries a poor prognosis with frequent relapses and high mortality. Curcumin is a yellow polyphenol compound with diverse pharmacological actions including anticancer, antioxidant, antidiabetic, anti-inflammatory, immunomodulatory, hepatoprotective, lipid-regulating, antidepressant, and antiarthritic. Many cellular and experimental studies have reported the benefits of curcumin in treating leukemia. Curcumin's anticancer effects are exerted via various mechanisms. Here, we review the effects of curcumin on various types of leukemia whilst considering its mechanisms of action.
Collapse
Affiliation(s)
- Hamideh Kouhpeikar
- Department of Hematology and Blood Bank, Cancer Molecular Pathology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Iran
| | - Alexandra E Butler
- Diabetes Research Center, Qatar Biomedical Research Institute, Doha, Qatar
| | - Faeze Bamian
- Department of Hematology and Blood Bank, Cancer Molecular Pathology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Iran
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | | | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
27
|
Orsini M, Morceau F, Dicato M, Diederich M. Autophagy as a pharmacological target in hematopoiesis and hematological disorders. Biochem Pharmacol 2018; 152:347-361. [PMID: 29656115 DOI: 10.1016/j.bcp.2018.04.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/10/2018] [Indexed: 12/14/2022]
Abstract
Autophagy is involved in many cellular processes, including cell homeostasis, cell death/survival balance and differentiation. Autophagy is essential for hematopoietic stem cell survival, quiescence, activation and differentiation. The deregulation of this process is associated with numerous hematological disorders and pathologies, including cancers. Thus, the use of autophagy modulators to induce or inhibit autophagy emerges as a potential therapeutic approach for treating these diseases and could be particularly interesting for differentiation therapy of leukemia cells. This review presents therapeutic strategies and pharmacological agents in the context of hematological disorders. The pros and cons of autophagy modulators in therapy will also be discussed.
Collapse
Affiliation(s)
- Marion Orsini
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Franck Morceau
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Marc Diederich
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
28
|
Magrì A, Reina S, De Pinto V. VDAC1 as Pharmacological Target in Cancer and Neurodegeneration: Focus on Its Role in Apoptosis. Front Chem 2018; 6:108. [PMID: 29682501 PMCID: PMC5897536 DOI: 10.3389/fchem.2018.00108] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 03/22/2018] [Indexed: 01/15/2023] Open
Abstract
Cancer and neurodegeneration are different classes of diseases that share the involvement of mitochondria in their pathogenesis. Whereas the high glycolytic rate (the so-called Warburg metabolism) and the suppression of apoptosis are key elements for the establishment and maintenance of cancer cells, mitochondrial dysfunction and increased cell death mark neurodegeneration. As a main actor in the regulation of cell metabolism and apoptosis, VDAC may represent the common point between these two broad families of pathologies. Located in the outer mitochondrial membrane, VDAC forms channels that control the flux of ions and metabolites across the mitochondrion thus mediating the organelle's cross-talk with the rest of the cell. Furthermore, the interaction with both pro-apoptotic and anti-apoptotic factors makes VDAC a gatekeeper for mitochondria-mediated cell death and survival signaling pathways. Unfortunately, the lack of an evident druggability of this protein, since it has no defined binding or active sites, makes the quest for VDAC interacting molecules a difficult tale. Pharmacologically active molecules of different classes have been proposed to hit cancer and neurodegeneration. In this work, we provide an exhaustive and detailed survey of all the molecules, peptides, and microRNAs that exploit VDAC in the treatment of the two examined classes of pathologies. The mechanism of action and the potential or effectiveness of each compound are discussed.
Collapse
Affiliation(s)
- Andrea Magrì
- Section of Molecular Biology, Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy.,Section of Biology and Genetics, Department of Biomedicine and Biotechnology, National Institute for Biomembranes and Biosystems, Section of Catania, Catania, Italy
| | - Simona Reina
- Section of Molecular Biology, Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy.,Section of Biology and Genetics, Department of Biomedicine and Biotechnology, National Institute for Biomembranes and Biosystems, Section of Catania, Catania, Italy
| | - Vito De Pinto
- Section of Biology and Genetics, Department of Biomedicine and Biotechnology, National Institute for Biomembranes and Biosystems, Section of Catania, Catania, Italy
| |
Collapse
|
29
|
Martínez-Castillo M, Villegas-Sepúlveda N, Meraz-Rios MA, Hernández-Zavala A, Berumen J, Coleman MA, Orozco L, Cordova EJ. Curcumin differentially affects cell cycle and cell death in acute and chronic myeloid leukemia cells. Oncol Lett 2018; 15:6777-6783. [PMID: 29616136 DOI: 10.3892/ol.2018.8112] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 01/24/2018] [Indexed: 02/07/2023] Open
Abstract
Curcumin is a phytochemical with potent anti-neoplastic properties. The antitumoral effects of curcumin in cells derived from chronic or acute myeloid leukemia have been already described. However, a comparative study of the cytostatic and cytotoxic effects of curcumin on chronic and acute myeloid leukemia cells has not yet been performed. In the present study, the cellular effects of curcumin on cell lines derived from chronic or acute myeloid leukemia were examined. Dose and time-response assays were performed with curcumin on HL-60 and K562 cells. Cell viability was evaluated with trypan blue exclusion test and cell death by flow cytometry using a fluorescent molecular probe. A cell cycle profile was analyzed, and protein markers of cell cycle progression and cell death were investigated. In the present study, the K562 cells showed a higher sensitivity to the cytostatic and cytotoxic effects of curcumin compared with HL-60. In addition, curcumin induced G1 phase arrest in HL-60 cells and G2/M phase arrest in K562 cells. Furthermore, curcumin-related cell death in HL-60 was associated with the processed forms of caspases-9 and -3 proteins, whereas in K562 cells, both the processed and the unprocessed forms were present. Accordingly, activity of these caspases was significantly higher in HL-60 cells compared with that in K562. In conclusion, curcumin elicits different cellular mechanisms in chronic or acute myeloid leukemia cells and the powerful antitumoral effect was more potent in K562 compared with HL-60 cells.
Collapse
Affiliation(s)
- Macario Martínez-Castillo
- Department of Molecular Biomedicine, Center of Studies and Advance Research, 07360 Mexico City, Mexico
| | | | - Marco A Meraz-Rios
- Department of Molecular Biomedicine, Center of Studies and Advance Research, 07360 Mexico City, Mexico
| | - Araceli Hernández-Zavala
- Section of Research and Postgraduate, Superior School of Medicine, National Institute Polytechnique, Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomás, 11350 Mexico City, Mexico
| | - Jaime Berumen
- Faculty of Medicine, National Autonomous University of Mexico (UNAM), AP, 04510 Mexico City, Mexico.,Unit of Genomic Medicine, Hospital General, 06720 Mexico City, Mexico
| | - Mathew A Coleman
- Lawrence Livermore National Laboratory, Livermore, CA 94550, USA.,Department of Radiation Oncology, University of California Davis, School of Medicine, Davis, CA 95817, USA
| | - Lorena Orozco
- National Institute of Genomic Medicine, Clinic Research, 14610 Mexico City, Mexico
| | - Emilio J Cordova
- National Institute of Genomic Medicine, Clinic Research, 14610 Mexico City, Mexico
| |
Collapse
|
30
|
Bisdemethoxycurcumin exerts pro-apoptotic effects in human pancreatic adenocarcinoma cells through mitochondrial dysfunction and a GRP78-dependent pathway. Oncotarget 2018; 7:83641-83656. [PMID: 27845899 PMCID: PMC5347794 DOI: 10.18632/oncotarget.13272] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 10/14/2016] [Indexed: 12/27/2022] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy, which is intrinsically resistant to current chemotherapies. Herein, we investigate whether bisdemethoxycurcumin (BDMC), a derivative of curcumin, potentiates gemcitabine in human pancreatic cancer cells. The result suggests that BDMC sensitizes gemcitabine by inducing mitochondrial dysfunctions and apoptosis in PANC-1 and MiaPaCa-2 pancreatic cancer cells. Utilizing two-dimensional gel electrophoresis and mass spectrometry, we identify 13 essential proteins with significantly altered expressions in response to gemcitabine alone or combined with BDMC. Protein-protein interaction network analysis pinpoints glucose-regulated protein 78 (GRP78) as the key hub activated by BDMC. We then reveal that BDMC upregulates GRP78 and facilitates apoptosis through eIF2α/CHOP pathway. Moreover, DJ-1 and prohibitin, two identified markers of chemoresistance, are increased by gemcitabine in PANC-1 cells. This could be meaningfully reversed by BDMC, suggesting that BDMC partially offsets the chemoresistance induced by gemcitabine. In summary, these findings show that BDMC promotes apoptosis through a GRP78-dependent pathway and mitochondrial dysfunctions, and potentiates the antitumor effect of gemcitabine in human pancreatic cancer cells.
Collapse
|
31
|
Mirzaei A, Ghaffari SH, Nikbakht M, Kamranzadeh Foumani H, Vaezi M, Mohammadi S, Alimoghaddam K, Ghavamzadeh A. OPN b and c Isoforms Doubtless Veto Anti-angiogenesis Effects of Curcumin in Combination with Conventional AML Regiment. Asian Pac J Cancer Prev 2017; 18:2591-2599. [PMID: 28952709 PMCID: PMC5720671 DOI: 10.22034/apjcp.2017.18.9.2591] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Osteopontin (OPN) is an extracellular structural protein that is secreted by osteoblasts and hematopoietic cells. It suppresses the proliferation of hematopoietic stem and also plays an important role in promoting survival and drug resistance in leukemic stem cells (LSCs). Since the role of OPN isoforms in AML angiogenesis are remaining controversial, in the present study, we aimed to evaluate whether curcumin (CUR), as a known natural component with anti-angiogenesis effects, in a combination of AML conventional regiment has the potency to preclude induced anti-angiogenesis effects of OPN isoforms or not? Leukemia cells were treated with different concentration of CUR and AML conventional drugs alone and/or in combination with together to find effective doses and IC50 values. Percentages of apoptotic cells were evaluated by Annexin/PI staining and mRNA levels of OPN isoforms and AKT/ VEGF-A and VEGF-C/ STAT3/ β-catenin/ CXCR4/ IL-6/ KDR gene expression were investigated by Real Time-PCR method. Moreover, to confirm OPN gene expression data, we investigated the effect of simvastatin and OPN siRNA as an OPN inhibitor on the cell proliferation and induction of apoptosis in the indicated cell lines. Our data display that Ara-c (2μM and 1μM in KG-1 and U937 cell lines respectively), CUR (40μM in both cell lines), and also their combination significantly increased the percentage of apoptotic cells. Moreover, the mRNA level of OPN isoforms were down regulated in the KG-1and U937 cell lines treated with Ara-c while, upregulated in KG-1and U937 cell lines treated with CUR and its combination. Our results suggest that despite anti-angiogenesis effects of CUR, AML cells probably evade from anti-angiogenesis effects of CUR via induction of OPN b and c isoform and related molecular pathways.
Collapse
Affiliation(s)
- Akram Mirzaei
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
The pan-Bcl2 Inhibitor AT101 Activates the Intrinsic Apoptotic Pathway and Causes DNA Damage in Acute Myeloid Leukemia Stem-Like Cells. Target Oncol 2017; 12:677-687. [DOI: 10.1007/s11523-017-0509-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
33
|
Addala E, Rafiei H, Das S, Bandy B, Das U, Karki SS, Dimmock JR. 3,5-Bis(3-dimethylaminomethyl-4-hydroxybenzylidene)-4-piperidone and related compounds induce glutathione oxidation and mitochondria-mediated cell death in HCT-116 colon cancer cells. Bioorg Med Chem Lett 2017; 27:3669-3673. [PMID: 28716495 DOI: 10.1016/j.bmcl.2017.07.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/03/2017] [Accepted: 07/04/2017] [Indexed: 11/15/2022]
Abstract
This study aims at investigating the cytotoxicity and some of the modes of action of 3,5-bis(3-dimethylamino-4-hydroxybenzylidene)-4-piperidone trihydrochloride 3 and two related compounds 2 (which lacks the dimethylaminomethyl groups) and 4 (which has an additional dimethylaminoethyl substituent in both aryl rings) in order to ascertain the contribution of dimethylaminoethyl substituent to bioactivity. The bioactivities of 2-4 were compared with curcumin 5. Both 2 and 3 displayed submicromolar GI50 values towards HCT-116 cells and were significantly more potent than 4, 5 and 5-fluorouracil (5-FU). All of the compounds displayed greater toxicity towards HCT-116 cells than human CRL-1790 non-malignant colon cells. In HCT-116 cells, the compounds 2, 3 and 5 increased the ratio of oxidised to reduced glutathione and destabilized the mitochondrial membrane potential. Both 2 and 5 produced an increase in mitochondrial superoxide and a burst in intracellular reactive oxygen species in HCT 116 cells. In addition, 2 and 4 stimulated respiration in rat liver mitochondria while 2 and 5 induced mitochondrial swelling. The results suggest that 2 and 5 cause oxidation or cross-linking of the thiols which control the mitochondrial permeability transition.
Collapse
Affiliation(s)
- Eshwari Addala
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon S7N 5E5, Canada
| | - Hossein Rafiei
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon S7N 5E5, Canada
| | - Swagatika Das
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon S7N 5E5, Canada
| | - Brian Bandy
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon S7N 5E5, Canada.
| | - Umashankar Das
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon S7N 5E5, Canada
| | - Subhas S Karki
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon S7N 5E5, Canada
| | - Jonathan R Dimmock
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon S7N 5E5, Canada.
| |
Collapse
|
34
|
Mirzaei A, Mohammadi S, Ghaffari SH, Nikbakht M, Bashash D, Alimoghaddam K, Ghavamzadeh A. Osteopontin b and c isoforms: Molecular Candidates Associated with Leukemic Stem Cell Chemoresistance in Acute Myeloid
Leukemia. Asian Pac J Cancer Prev 2017; 18:1707-1715. [PMID: 28670893 PMCID: PMC6373801 DOI: 10.22034/apjcp.2017.18.6.1707] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Despite impressive advances in therapeutic approaches, long-term survival with acute myeloid leukemia (AML) is
low as a result of treatment resistance and frequent relapse. Among multitude oncogenic proteins involved in acquisition
of a chemo-resistanr phenotype, osteopontin (OPN) recently has attracted marked attention. In spite of the well-defined
association between OPN expression and cure rate with solid tumors, there is a scarcity of information on any role of this
protein in AML cases. Based on the critical role of OPN in cell survival, it seems reasonable to hypothesize that isoform
expression levels may impact on regulation of apoptosis in AML cells in response to conventional chemotherapeutic
drugs and its relation to relapse. To investigate associations between induction of apoptosis and OPN isoform expression,
two distinct AML cell lines (KG-1 as a leukemic stem cell model and U937) were treated with chemotherapy drugs,
and cell viability and apoptosis were evaluated by MTT and Annexin/PI assay. After determination of appropriate drug
doses, mRNA expression levels of OPN isoforms and OPN-related genes were investigated. Our results demonstrated
for the first time that acquired up-regulation of OPN-b and c isoforms might prevent conventional chemotherapy
regimen-induced apoptosis in AML cells. Moreover, upregulation of OPN-b and c in AML cells appears concurrent
with upregulation of AKT/VEGF/CXCR4/STAT3/ IL-6 gene expression. To sum up, this study suggests that OPN-b
and c isoforms could be considered as unique beneficial molecular biomarkers associated with leukemic stem cell
chemoresistance. Hence, they have potential as molecular candidates for detection of minimal residual disease (MRD)
and determination of remission in AML patients. Further evaluation with quantative real time PCR on patient samples
for confirmation appears warranted.
Collapse
Affiliation(s)
- Akram Mirzaei
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran. ,
| | | | | | | | | | | | | |
Collapse
|
35
|
Kumari M, Ray L, Purohit MP, Patnaik S, Pant AB, Shukla Y, Kumar P, Gupta KC. Curcumin loading potentiates the chemotherapeutic efficacy of selenium nanoparticles in HCT116 cells and Ehrlich's ascites carcinoma bearing mice. Eur J Pharm Biopharm 2017; 117:346-362. [PMID: 28499854 DOI: 10.1016/j.ejpb.2017.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 04/28/2017] [Accepted: 05/06/2017] [Indexed: 12/20/2022]
Abstract
The anticancer properties of selenium (Se) and curcumin nanoparticles in solo formulations as well as in combination with other therapeutic agents have been proved time and again. Exploiting this facet of the two, we clubbed their tumoricidal characteristics and designed curcumin loaded Se nanoparticles (Se-CurNPs) to achieve an enhanced therapeutic effect. We evaluated their therapeutic effects on different cancer cell lines and Ehrlich's ascites carcinoma mouse model. In vitro results showed that Se-CurNPs were most effective on colorectal carcinoma cells (HCT116) compared to the other cancer cell lines used and possessed pleiotropic anticancer effects. The therapeutic effect on HCT116 was primarily attributed to an elevated level of autophagy and apoptosis as evident from significant up-regulation of autophagy associated (LC3B-II) and pro-apoptotic (Bax) proteins, down-regulation of anti-apoptotic (Bcl-2) protein and Cytochrome c (cyt c) release from mitochondria along with reduced NFκB signaling and EMT based machineries marked by downregulation of inflammation (NFκB, phospho-NFκB) and epithelial-mesenchymal transition (CD44, N-cadherin) associated proteins. In vivo studies on Ehrlich's ascites carcinoma (EAC) mice model indicated that Se-CurNPs significantly reduced the tumor load and enhanced the mean survival time (days) of tumor-bearing EAC mice.
Collapse
Affiliation(s)
- Manisha Kumari
- CSIR-Institute of Genomics and Integrative Biology, Delhi University Campus, Mall Road, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - L Ray
- CSIR-Indian Institute of Toxicology Research, M.G. Marg, Lucknow 226 001, Uttar Pradesh, India
| | - M P Purohit
- CSIR-Indian Institute of Toxicology Research, M.G. Marg, Lucknow 226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - S Patnaik
- CSIR-Indian Institute of Toxicology Research, M.G. Marg, Lucknow 226 001, Uttar Pradesh, India
| | - A B Pant
- CSIR-Indian Institute of Toxicology Research, M.G. Marg, Lucknow 226 001, Uttar Pradesh, India
| | - Y Shukla
- CSIR-Indian Institute of Toxicology Research, M.G. Marg, Lucknow 226 001, Uttar Pradesh, India
| | - P Kumar
- CSIR-Institute of Genomics and Integrative Biology, Delhi University Campus, Mall Road, Delhi 110007, India
| | - K C Gupta
- CSIR-Institute of Genomics and Integrative Biology, Delhi University Campus, Mall Road, Delhi 110007, India; CSIR-Indian Institute of Toxicology Research, M.G. Marg, Lucknow 226 001, Uttar Pradesh, India; Department of Biological Sciences and Bioengineering (BSBE) and Centre for Environmental Science and Engineering (CESE), Indian Institute of Technology, Kanpur, India.
| |
Collapse
|
36
|
Owen HC, Appiah S, Hasan N, Ghali L, Elayat G, Bell C. Phytochemical Modulation of Apoptosis and Autophagy: Strategies to Overcome Chemoresistance in Leukemic Stem Cells in the Bone Marrow Microenvironment. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 135:249-278. [PMID: 28807161 DOI: 10.1016/bs.irn.2017.02.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Advances in scientific research and targeted treatment regimes have improved survival rates for many cancers over the past few decades. However, for some types of leukemia, including acute lymphoblastic and acute myeloid leukemia, mortality rates have continued to rise, with chemoresistance in leukemic stem cells (LSCs) being a major contributing factor. Most cancer drug therapies act by inducing apoptosis in dividing cells but are ineffective in targeting quiescent LSCs. Niches in the bone marrow, known as leukemic niches, behave as "sanctuaries" where LSCs acquire drug resistance. This review explores the role of the bone marrow environment in the maintenance of LSCs and its contribution to chemoresistance and considers current research on the potential use of phytochemicals to overcome chemoresistance through the modulation of signaling pathways involved in the survival and death of leukemic clonal cells and/or leukemic stem cells. Phytochemicals from traditional Chinese medicine, namely baicalein, chrysin, wogonin (constituents of Scutellaria baicalensis; huáng qín; ), curcumin (a constituent of Curcuma longa, jiāng huáng, ), and resveratrol (a constituent of Polygonum cuspidatum; hŭ zhàng, ) have been shown to induce apoptosis in leukemic cell lines, with curcumin and resveratrol also causing cell death via the induction of autophagy (a nonapoptotic pathway). In order to be effective in eliminating LSCs, it is important to target signaling pathways (such as Wnt/β-catenin, Notch, and Hedgehog). Resveratrol has been reported to induce apoptosis in leukemic cells through the inhibition of the Notch and Sonic hedgehog signaling pathways, therefore showing potential to affect LSCs. While these findings are of interest, there is a lack of reported research on the modulatory effect of phytochemicals on the autophagic cell death pathway in leukemia, and on the signaling pathways involved in the maintenance of LSCs, highlighting the need for further work in these areas.
Collapse
Affiliation(s)
- Helen C Owen
- Faculty of Science and Technology, Middlesex University, The Burroughs, Hendon, London, United Kingdom.
| | - Sandra Appiah
- Faculty of Science and Technology, Middlesex University, The Burroughs, Hendon, London, United Kingdom.
| | - Noor Hasan
- Faculty of Science and Technology, Middlesex University, The Burroughs, Hendon, London, United Kingdom
| | - Lucy Ghali
- Faculty of Science and Technology, Middlesex University, The Burroughs, Hendon, London, United Kingdom
| | - Ghada Elayat
- Faculty of Science and Technology, Middlesex University, The Burroughs, Hendon, London, United Kingdom
| | - Celia Bell
- Faculty of Science and Technology, Middlesex University, The Burroughs, Hendon, London, United Kingdom
| |
Collapse
|
37
|
Zahed Panah M, Nikbakht M, Sajjadi SM, Rostami S, Norooznezhad AH, Kamranzadeh Fumani H, Ghavamzadeh A, Mohammadi S. Anti-Apoptotic Effects of Osteopontin via the Up-Regulation of AKT/mTOR/β-Catenin Loop in Acute Myeloid Leukemia Cells. Int J Hematol Oncol Stem Cell Res 2017; 11:148-157. [PMID: 28875010 PMCID: PMC5574411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background: The conventional chemotherapeutic regimens which applied for treatment of acute myeloid leukemia (AML) mostly target tumor bulk but not leukemic stem cells (LSCs). Aberrant expression or activation of mediators such as osteopontin (OPN) or PI3K/PTEN/Akt/mTOR pathway plays a key role in making prone to develop leukemia. Preventing or treating cancer by curcumin (CUR) has been suggested recently. CUR induces apoptosis and growth inhibition through various mechanisms in leukemic cells. In present study, we tried to measure the toxic response in vitro to CUR for evaluation ofchangesin cell viability, survival and molecular-mediated resistance in primary AML cells. Materials and Methods: Isolated primary CD34+/CD38- bone marrow derived AML cells were treated with CUR, Daunorubicin (DNR) and/or their combination by MTT assay, Annexin V/PI staining, and colony-formation. The mRNA expression of OPN/AKT/mTOR/PTEN/β-catenin genes was measured by Real-Time PCR. The siRNA against OPN was applied for CUR- treated cells. Results: Growth inhibition effect of DNR increased in combination with CUR on primary CD34+/CD38- AML cells. Suppression of OPN with siRNA increased the cytotoxic effects of CUR. Likewise, OPN gene expression increased in response to CUR treatment in AML cells. AKT, mTOR, β-catenin or PTEN gene expression increased by CUR, but OPN siRNA decreased the level of mRNA expression of mentioned molecular pathway. Conclusion: The chemo-resistance of AML cells against therapy might be relevant to increasing of OPN mRNA expression and activity of other mediators including AKT, mTOR, PTEN, and β-catenin. In this context, targeting of OPN might be more impact on CD34+ AML cells.
Collapse
Affiliation(s)
- Mahdi Zahed Panah
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohsen Nikbakht
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mehdi Sajjadi
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Shahrbano Rostami
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hosein Kamranzadeh Fumani
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ardeshir Ghavamzadeh
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Mohammadi
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Hamdy R, Ziedan NI, Ali S, Bordoni C, El-Sadek M, Lashin E, Brancale A, Jones AT, Westwell AD. Synthesis and evaluation of 5-(1H-indol-3-yl)-N-aryl-1,3,4-oxadiazol-2-amines as Bcl-2 inhibitory anticancer agents. Bioorg Med Chem Lett 2016; 27:1037-1040. [PMID: 28087272 DOI: 10.1016/j.bmcl.2016.12.061] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/22/2016] [Accepted: 12/24/2016] [Indexed: 11/18/2022]
Abstract
A series of 5-(1H-indol-3-yl)-N-aryl-1,3,4-oxadiazol-2-amines 8a-j has been designed, synthesized and tested in vitro as potential pro-apoptotic Bcl-2-inhibitory anticancer agents based on our previous lead compound 8a. Synthesis of the target compounds was readily accomplished through a cyclisation reaction between indole-3-carboxylic acid hydrazide (5) and substituted isothiocyanates 6a-j, followed by oxidative cyclodesulfurization of the corresponding thiosemicarbazide 7a-j using 1,3-dibromo-5,5-dimethylhydantoin. Active compounds of the series 8a-j were found to have sub-micromolar IC50 values selectively in Bcl-2 expressing human cancer cell lines; notably the 2-nitrophenyl analogue 8a was found to exhibit potent activity, and compounds 8a and 8e possessed comparable Bcl-2 binding affinity (ELISA assay) to the established natural product-based Bcl-2 inhibitor, gossypol. Molecular modeling studies helped to further rationalise anti-apoptotic Bcl-2 binding, and identified compounds 8a and 8e as candidates for further development as Bcl-2 inhibitory anticancer agents.
Collapse
Affiliation(s)
- Rania Hamdy
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, Wales, UK; Faculty of Pharmacy, Zagazig University, Egypt
| | - Noha I Ziedan
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, Wales, UK; Faculty of Pharmacy, Zagazig University, Egypt
| | - Samia Ali
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, Wales, UK; National Research Centre, Cairo, Egypt
| | - Cinzia Bordoni
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, Wales, UK
| | | | | | - Andrea Brancale
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, Wales, UK
| | - Arwyn T Jones
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, Wales, UK
| | - Andrew D Westwell
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, Wales, UK.
| |
Collapse
|
39
|
Popova AA, Depew C, Permana KM, Trubitsyn A, Peravali R, Ordiano JÁG, Reischl M, Levkin PA. Evaluation of the Droplet-Microarray Platform for High-Throughput Screening of Suspension Cells. SLAS Technol 2016; 22:163-175. [PMID: 28095175 DOI: 10.1177/2211068216677204] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Phenotypic cell-based high-throughput screenings play a central role in drug discovery and toxicology. The main tendency in cell screenings is the increase of the throughput and decrease of reaction volume in order to accelerate the experiments, reduce the costs, and enable screenings of rare cells. Conventionally, cell-based assays are performed in microtiter plates, which exist in 96- to 1536-wells formats and cannot be further miniaturized. In addition, performing screenings of suspension cells is associated with risk of losing cell content during the staining procedures and incompatibility with high-content microscopy. Here, we evaluate the Droplet-Microarray screening platform for culturing, screening, and imaging of suspension cells. We demonstrate pipetting-free cell seeding and proliferation of cells in individual droplets of 3-80 nL in volume. We developed a methodology to perform parallel treatment, staining, and fixation of suspension cells in individual droplets. Automated imaging of live suspension cells directly in the droplets combined with algorithms for pattern recognition for image analysis is demonstrated. We evaluated the developed methodology by performing a dose-response study with antineoplastic drugs. We believe that the DMA screening platform carries great potential to be adopted for broad spectrum of screenings of suspension cells.
Collapse
Affiliation(s)
- Anna A Popova
- 1 Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Eggenstein-Leopoldshafen, Germany
| | - Claire Depew
- 1 Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Eggenstein-Leopoldshafen, Germany
| | - Katya Manuella Permana
- 1 Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Eggenstein-Leopoldshafen, Germany
| | - Alexander Trubitsyn
- 1 Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Eggenstein-Leopoldshafen, Germany
| | - Ravindra Peravali
- 1 Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Eggenstein-Leopoldshafen, Germany
| | | | - Markus Reischl
- 2 Karlsruhe Institute of Technology, Institute for Applied Computer Science, Eggenstein-Leopoldshafen, Germany
| | - Pavel A Levkin
- 1 Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Eggenstein-Leopoldshafen, Germany.,3 Karlsruhe Institute of Technology, Institute of Organic Chemistry, Karlsruhe, Germany
| |
Collapse
|
40
|
Tima S, Okonogi S, Ampasavate C, Pickens C, Berkland C, Anuchapreeda S. Development and Characterization of FLT3-Specific Curcumin-Loaded Polymeric Micelles as a Drug Delivery System for Treating FLT3-Overexpressing Leukemic Cells. J Pharm Sci 2016; 105:3645-3657. [PMID: 27751588 DOI: 10.1016/j.xphs.2016.09.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/30/2016] [Accepted: 09/09/2016] [Indexed: 01/04/2023]
Abstract
This study aimed at developing a curcumin (CM) nanoparticle targeted to Feline McDonough Sarcoma (FMS)-like tyrosine kinase 3 (FLT3) protein on the surface of leukemic cells and at evaluating their properties, specificity, cytotoxicity, and inhibitory effect on FLT3 protein level in FLT3-overexpressing leukemic cells, EoL-1, and MV-4-11 cells. FLT3-specific peptides were conjugated onto modified poloxamer 407 using the copper-catalyzed azide-alkyne cycloaddition reaction. The thin film hydration method was performed for FLT3-specific CM-loaded polymeric micelles (FLT3-CM-micelles) preparation. Flow cytometry and fluorescence microscopy were used to determine rate of cellular uptake. 3-(4,5-dimethythiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay was used to test the cytotoxicity of the micelles on leukemic cells. FLT3-CM-micelles demonstrated a mean particle size less than 50 nm, high entrapment efficiency, and high rate of CM uptake by leukemic cells. The intracellular CM fluorescence is related to FLT3 protein levels on the leukemic cell surfaces. Moreover, FLT3-CM-micelles demonstrated an excellent cytotoxic effect and decreased FLT3 protein expression in the leukemic cells. The FLT3-CM-micelles could enhance both solubility and cytotoxicity of CM on FLT3-overexpressing leukemic cells. These promising nanoparticles may be used for enhancing antileukemic activity of CM and developed as a targeted drug delivery system in the future.
Collapse
Affiliation(s)
- Singkome Tima
- Nanoscience and Nanotechnology Program, Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn Okonogi
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chadarat Ampasavate
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chad Pickens
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Kansas 66047
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Kansas 66047.
| | - Songyot Anuchapreeda
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
41
|
Pimentel-Gutiérrez HJ, Bobadilla-Morales L, Barba-Barba CC, Ortega-De-La-Torre C, Sánchez-Zubieta FA, Corona-Rivera JR, González-Quezada BA, Armendáriz-Borunda JS, Silva-Cruz R, Corona-Rivera A. Curcumin potentiates the effect of chemotherapy against acute lymphoblastic leukemia cells via downregulation of NF-κB. Oncol Lett 2016; 12:4117-4124. [PMID: 27895780 DOI: 10.3892/ol.2016.5217] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 05/16/2016] [Indexed: 11/06/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) accounts for 30% of all pediatric cancers. Currently available treatments exhibit toxicity and certain patients may develop resistance. Thus, less toxic and chemoresistance-reversal agents are required. In the present study, the potential effect of curcumin, a component of Curcuma longa, as a pharmacological co-adjuvant of several chemotherapeutic agents against ALL, including prednisone, 6-mercaptopurine, dexamethasone, cyclophosphamide, l-asparaginase, vincristine, daunorubicin, doxorubicin, methotrexate and cytarabine, was investigated in the REH ALL cell line cultures treated in combination with chemotherapeutic agents and curcumin. The results of cell viability, gene expression and activation of NF-κB and caspase 3 indicated that curcumin potentiates the anticancer effects of the aforementioned chemotherapeutic agents in the REH ALL cell line. Following treatment with the above chemotherapeutic agents, curcumin enhanced caspase-3 activation and downregulated nuclear factor-kappa B (NF-κB) activation. Curcumin also downregulated the oxidative stress induced by certain chemotherapies. Notably, curcumin did not affect the gene expression of cell survival proteins such as B-cell lymphoma (Bcl)-2, Bcl-extra large, survivin, c-Myc and cyclin D1, which are regulated by the NF-κB transcription factor. In conclusion, curcumin has the potential to improve the effect of chemotherapeutic agents against ALL.
Collapse
Affiliation(s)
- Helia Judith Pimentel-Gutiérrez
- Cytogenetics, Genotoxicity and Biomonitoring Laboratory, Human Genetic Institute 'Dr. Enrique Corona Rivera', PhD Program in Molecular Biology, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco 44340, México; Cytogenetics Unit, Pediatric Hematology and Oncology Service, Pediatric Division, Civil Hospital of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Lucina Bobadilla-Morales
- Cytogenetics, Genotoxicity and Biomonitoring Laboratory, Human Genetic Institute 'Dr. Enrique Corona Rivera', PhD Program in Molecular Biology, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco 44340, México; Cytogenetics Unit, Pediatric Hematology and Oncology Service, Pediatric Division, Civil Hospital of Guadalajara, Guadalajara, Jalisco 44340, México
| | - César Cenobio Barba-Barba
- Cytogenetics Unit, Pediatric Hematology and Oncology Service, Pediatric Division, Civil Hospital of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Citlalli Ortega-De-La-Torre
- Cytogenetics Unit, Pediatric Hematology and Oncology Service, Pediatric Division, Civil Hospital of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Fernando Antonio Sánchez-Zubieta
- Cytogenetics Unit, Pediatric Hematology and Oncology Service, Pediatric Division, Civil Hospital of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Jorge Román Corona-Rivera
- Cytogenetics, Genotoxicity and Biomonitoring Laboratory, Human Genetic Institute 'Dr. Enrique Corona Rivera', PhD Program in Molecular Biology, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco 44340, México; Cytogenetics Unit, Pediatric Hematology and Oncology Service, Pediatric Division, Civil Hospital of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Betsy Annel González-Quezada
- Cytogenetics, Genotoxicity and Biomonitoring Laboratory, Human Genetic Institute 'Dr. Enrique Corona Rivera', PhD Program in Molecular Biology, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Juan S Armendáriz-Borunda
- Molecular Biology and Gene Therapy Institute, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Rocío Silva-Cruz
- Cytogenetics, Genotoxicity and Biomonitoring Laboratory, Human Genetic Institute 'Dr. Enrique Corona Rivera', PhD Program in Molecular Biology, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Alfredo Corona-Rivera
- Cytogenetics, Genotoxicity and Biomonitoring Laboratory, Human Genetic Institute 'Dr. Enrique Corona Rivera', PhD Program in Molecular Biology, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco 44340, México; Cytogenetics Unit, Pediatric Hematology and Oncology Service, Pediatric Division, Civil Hospital of Guadalajara, Guadalajara, Jalisco 44340, México
| |
Collapse
|
42
|
Mohammadi S, Ghaffari SH, Shaiegan M, Nikogoftar Zarif M, Nikbakht M, Alimoghaddam K, Ghavamzadeh A. Curcumin Veto the Effects of Osteopontin (OPN) Specific Inhibitor on Leukemic Stem Cell Colony Forming Potential via Promotion of OPN Overexpression. Int J Hematol Oncol Stem Cell Res 2016; 10:120-9. [PMID: 27489587 PMCID: PMC4969556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is an immunophenotypically heterogeneous malignant disease, in which CD34 positivity is associated with poor prognosis. Osteopontin (OPN) plays different roles in physiologic and pathologic conditions like: survival, metastasis and cell protection from cytotoxic and apoptotic stimuli. Due to anti-apoptotic effect of OPN in normal and malignant cells, silencing of OPN leads to elevation of sensitivity towards chemotherapeutic agents and attenuates cancer cells migration and invasion. Therefore, the aim of this study was to evaluate OPN roles in modulating curcumin-mediated growth inhibitory on leukemic stem cells (LSCs) colony forming potential and survival in AML cell lines and primary CD34+/CD38- bone marrow-derived AML cells. MATERIALS AND METHODS Primary human CD34+/CD38- cells were isolated from bone marrow mononuclear cells of 10 AML patients at initial state of diagnosis, using a CD34 Multi sort kit. The growth inhibitory effects of curcumin (CUR) were evaluated by MTT and colony-formation assays. Apoptosis was analyzed by 7AAD assay in CD34+ KG-1, U937 cell lines and primary isolated cells. Short interfering RNA (siRNA) against OPN was used for OPN silencing in both cell lines and primary AML cells. Then, transfected cells were incubated with/without curcumin. The change in OPN gene expression was examined by Real-time PCR. RESULTS CUR inhibited proliferation and induced apoptosis in both KG-1 and U937 cells and also primary isolated AML cells. OPN silencing by siRNA increased the susceptibility of KG-1, U937 and primary CD34+/CD38- AML cells to apoptosis. Moreover, soft agar colony assays revealed that silencing of OPN with siRNA significantly decreased colony numbers in LSCs compared with the non-targeting group. Furthermore, CD34+/CD38- populations as a main LSCs compartment through OPN overexpression towards CUR treatment might be nullified the inhibitory effects of OPN siRNA on their survival and colony forming potential. CONCLUSION Taken together, our results suggested that knockdown of OPN using OPN specific siRNA significantly decreased colony numbers in LSCs and this effect might be vetoed by LSCs via induction of OPN overexpressionin combination of CUR and siRNA.
Collapse
Affiliation(s)
- Saeed Mohammadi
- Iranian Blood Transfusion Research Center, High Institute for Education and Research in Transfusion Medicine, Tehran, Iran,Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed H. Ghaffari
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojgan Shaiegan
- Iranian Blood Transfusion Research Center, High Institute for Education and Research in Transfusion Medicine, Tehran, Iran
| | - Mahin Nikogoftar Zarif
- Iranian Blood Transfusion Research Center, High Institute for Education and Research in Transfusion Medicine, Tehran, Iran
| | - Mohsen Nikbakht
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kamran Alimoghaddam
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ardeshir Ghavamzadeh
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Zhou W, Xu J, Gelston E, Wu X, Zou Z, Wang B, Zeng Y, Wang H, Liu A, Xu L, Liu Q. Inhibition of Bcl-xL overcomes polyploidy resistance and leads to apoptotic cell death in acute myeloid leukemia cells. Oncotarget 2016; 6:21557-71. [PMID: 26188358 PMCID: PMC4673286 DOI: 10.18632/oncotarget.4306] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 05/13/2015] [Indexed: 11/28/2022] Open
Abstract
Small molecular inhibitors or drugs targeting specific molecular alterations are widely used in clinic cancer therapy. Despite the success of targeted therapy, the development of drug resistance remains a challenging problem. Identifying drug resistance mechanisms for targeted therapy is an area of intense investigation, and recent evidence indicates that cellular polyploidy may be involved. Here, we demonstrate that the cell cycle kinase inhibitor, Oxindole-1 (Ox-1), induces mitotic slippage, causing resistant polyploidy in acute myeloid leukemia (AML) cells. Indeed, Ox-1 decreases the kinase activity of CDK1 (CDC2)/cyclin B1, leading to inhibition of Bcl-xL phosphorylation and subsequent resistance to apoptosis. Addition of ABT-263, a Bcl-2 family inhibitor, to Ox-1, or the other polyploidy-inducer, ZM447439 (ZM), produces a synergistic loss of cell viability with greater sustained tumor growth inhibition in AML cell lines and primary AML blasts. Furthermore, genetic knockdown of Bcl-xL, but not Bcl-2, exhibited synergistic inhibition of cell growth in combination with Ox-1 or ZM. These data demonstrate that Bcl-xL is a key factor in polyploidization resistance in AML, and that suppression of Bcl-xL by ABT-263, or siRNAs, may hold therapeutic utility in drug-resistant polyploid AML cells.
Collapse
Affiliation(s)
- Weihua Zhou
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, China.,Department of Oncology, the Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Jie Xu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, China.,Department of Oncology, the Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Elise Gelston
- University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Xing Wu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Zhengzhi Zou
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Bin Wang
- Department of Ultrasound, Union Hospital, Tongji Medical Collage of Huazhong University of Science and Technology, Wuhan, China
| | - Yunxin Zeng
- Department of Hematology, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hua Wang
- Department of Hematological Oncology, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Anwen Liu
- Department of Oncology, the Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Lingzhi Xu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Quentin Liu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, China.,Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| |
Collapse
|
44
|
Bollu VS, Barui AK, Mondal SK, Prashar S, Fajardo M, Briones D, Rodríguez-Diéguez A, Patra CR, Gómez-Ruiz S. Curcumin-loaded silica-based mesoporous materials: Synthesis, characterization and cytotoxic properties against cancer cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 63:393-410. [DOI: 10.1016/j.msec.2016.03.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 02/28/2016] [Accepted: 03/01/2016] [Indexed: 01/22/2023]
|
45
|
Dos S Beirigo PJ, Torquato HFV, Dos Santos CHC, Carvalho MGD, Castro RN, Paredes-Gamero EJ, de Sousa PT, Jacinto MJ, da Silva VC. [1-8-NαC]-Zanriorb A1, a Proapoptotic Orbitide from Leaves of Zanthoxylum riedelianum. JOURNAL OF NATURAL PRODUCTS 2016; 79:1454-8. [PMID: 27082849 DOI: 10.1021/acs.jnatprod.5b00177] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
A new orbitide named [1-8-NαC]-zanriorb A1 (1) was isolated and characterized from the leaves of Zanthoxylum riedelianum using NMR and mass spectrometry. The absolute configuration of the amino acids was determined using Marfey's method on the acid hydrolysates. Compound 1 induced cell death by apoptosis in Jurkat leukemia T cells (IC50 218 nM).
Collapse
Affiliation(s)
- Pâmela J Dos S Beirigo
- Departamento de Química, Universidade Federal de Mato Grosso , Cuiabá, MT 78060-900, Brazil
| | - Heron F V Torquato
- Departamento de Bioquímica, Universidade Federal de São Paulo , São Paulo, SP 04039-032, Brazil
| | - Carlos H C Dos Santos
- Departamento de Química, Universidade Federal Rural do Rio de Janeiro , Seropédica, RJ 23890-000, Brazil
| | - Mário G de Carvalho
- Departamento de Química, Universidade Federal Rural do Rio de Janeiro , Seropédica, RJ 23890-000, Brazil
| | - Rosane N Castro
- Departamento de Química, Universidade Federal Rural do Rio de Janeiro , Seropédica, RJ 23890-000, Brazil
| | - Edgar J Paredes-Gamero
- Departamento de Bioquímica, Universidade Federal de São Paulo , São Paulo, SP 04039-032, Brazil
| | - Paulo T de Sousa
- Departamento de Química, Universidade Federal de Mato Grosso , Cuiabá, MT 78060-900, Brazil
| | - Marcos J Jacinto
- Departamento de Química, Universidade Federal de Mato Grosso , Cuiabá, MT 78060-900, Brazil
| | - Virgínia C da Silva
- Departamento de Química, Universidade Federal de Mato Grosso , Cuiabá, MT 78060-900, Brazil
| |
Collapse
|
46
|
Papież MA, Krzyściak W, Szade K, Bukowska-Straková K, Kozakowska M, Hajduk K, Bystrowska B, Dulak J, Jozkowicz A. Curcumin enhances the cytogenotoxic effect of etoposide in leukemia cells through induction of reactive oxygen species. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:557-70. [PMID: 26893544 PMCID: PMC4745860 DOI: 10.2147/dddt.s92687] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Curcumin may exert a more selective cytotoxic effect in tumor cells with elevated levels of free radicals. Here, we investigated whether curcumin can modulate etoposide action in myeloid leukemia cells and in normal cells of hematopoietic origin. HL-60 cell line, normal myeloid progenitor cluster of differentiation (CD)-34+ cells, and granulocytes were incubated for 4 or 24 hours at different concentrations of curcumin and/or etoposide. Brown Norway rats with acute myeloid leukemia (BNML) were used to prove the influence of curcumin on etoposide action in vivo. Rats were treated with curcumin for 23 days and etoposide was administered for the final 3 days of the experiment. Curcumin synergistically potentiated the cytotoxic effect of etoposide, and it intensified apoptosis and phosphorylation of the histone H2AX induced by this cytostatic drug in leukemic HL-60 cells. In contrast, curcumin did not significantly modify etoposide-induced cytotoxicity and H2AX phosphorylation in normal CD34+ cells and granulocytes. Curcumin modified the cytotoxic action of etoposide in HL-60 cells through intensification of free radical production because preincubation with N-acetyl-l-cysteine (NAC) significantly reduced the cytotoxic effect of curcumin itself and a combination of two compounds. In contrast, NAC did not decrease the cytotoxic effect of etoposide. Thus, oxidative stress plays a greater role in the cytotoxic effect of curcumin than that of etoposide in HL-60 cells. In vitro results were confirmed in a BNML model. Pretreatment with curcumin enhanced the antileukemic activity of etoposide in BNML rats (1.57-fold tumor reduction versus etoposide alone; P<0.05) and induced apoptosis of BNML cells more efficiently than etoposide alone (1.54-fold change versus etoposide alone; P<0.05), but this treatment protected nonleukemic B-cells from apoptosis. Thus, curcumin can increase the antileukemic effect of etoposide through reactive oxygen species in sensitive myeloid leukemia cells, and it is harmless to normal human cells.
Collapse
Affiliation(s)
- Monika A Papież
- Department of Cytobiology, Jagiellonian University Medical College, Krakow, Poland
| | - Wirginia Krzyściak
- Department of Medical Diagnostic, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Krzysztof Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Karolina Bukowska-Straková
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland; Department of Clinical Immunology, Institute of Pediatrics, Krakow, Poland
| | - Magdalena Kozakowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Karolina Hajduk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Beata Bystrowska
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland; Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
47
|
Targeting CXCR4/SDF-1 axis by lipopolymer complexes of siRNA in acute myeloid leukemia. J Control Release 2016; 224:8-21. [DOI: 10.1016/j.jconrel.2015.12.052] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/22/2015] [Accepted: 12/28/2015] [Indexed: 02/07/2023]
|
48
|
Rao J, Li F, Zhang RY, Zhou HH, Chen GA. BH3 mimetic ABT-737 induces apoptosis in CD34 + acute myeloid leukemia cells and shows synergistic effect with conventional chemotherapeutic drugs. Asia Pac J Clin Oncol 2015; 13:e144-e152. [PMID: 26552712 DOI: 10.1111/ajco.12420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2015] [Indexed: 11/27/2022]
Abstract
AIMS Acute myeloid leukemia (AML) is an immunophenotypically heterogenous malignant disease. The early immature CD34+ AML cell subpopulation is frequently impervious to intensive chemotherapy, making them largely responsible for relapse of AML. CD34+ AML cells have higher level of Bcl-2 protein expression than the CD34- subpopulation. As such, development of drugs that specifically target the Bcl-2 may have the potential to eliminate immature CD34+ AML progenitor cells and provide therapeutic benefit. In this work, we made an attempt to investigate the cytotoxic effect of a novel Bcl-2 family inhibitor, ABT-737, on CD34+ AML cell lines (KG1a and Kasumi-1) as well as CD34+ primary AML cells. METHODS Primary human CD34+ cells were isolated from bone marrow mononuclear cells using CD34 MicroBead kit. The growth inhibitory effect was measured by cell counting kit-8. Apoptosis was analyzed by annexin V/PI assays. Protein expression was determined by Western blotting analysis. RESULTS Inhibition of Bcl-2 by ABT-737 effectively inhibited growth and induced apoptosis in CD34+ AML cell lines and CD34+ primary AML cells without affecting CD34+ normal hematopoietic cells. Furthermore, Western blot analysis showed that ABT-737 induced apoptosis associated with caspase-3 activation and poly ADP-ribose polymerase (PARP) degradation. Finally, ABT-737 synergistically enhanced the cytotoxic effect of cytarabine and daunorubicin in CD34+ AML cells. CONCLUSION Taken together, these findings indicate that ABT-737 may offer as a promising molecular targeting agent in CD34+ AML.
Collapse
Affiliation(s)
- Jia Rao
- Department of Hematology, First Affiliated Hospital, NanChang University, Nanchang, China
| | - Fei Li
- Department of Hematology, First Affiliated Hospital, NanChang University, Nanchang, China
| | - Rong-Yan Zhang
- Department of Hematology, First Affiliated Hospital, NanChang University, Nanchang, China
| | - Huan-Huan Zhou
- Department of Hematology, First Affiliated Hospital, NanChang University, Nanchang, China
| | - Guo-An Chen
- Department of Hematology, First Affiliated Hospital, NanChang University, Nanchang, China
| |
Collapse
|
49
|
Liu B, Li Y. Impact of Conditional miRNA126 Overexpression on Apoptosis-Resistant Endothelial Cell Production. PLoS One 2015; 10:e0126661. [PMID: 25961846 PMCID: PMC4427270 DOI: 10.1371/journal.pone.0126661] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 04/06/2015] [Indexed: 01/08/2023] Open
Abstract
The activation of endothelial cells is essential to repair damage caused by atherosclerosis via endothelial cell proliferation and migration. Overexpression of VEGF (vascular endothelial growth factor) and the downstream gene, B-cell lymphoma-2 (BCL-2) could result in apoptosis-resistant endothelial cells, which are responsible for aggravated hyperplasia and instable plaques generation. Previous studies have shown that miRNA126 could regulate the expression of VEGF. Here, we verified the existence of a miRNA126 binding site in VEGF's 3'UTR. Additionally, VEGF regulated BCL-2 expression via AP1 (Activator Protein 1) binding site in BCL-2's promoter. Next, we established an apoptosis-resistant endothelial cell line and constructed a lentiviral vector to express miRNA126 under the control of the BCL-2 promoter to investigate whether conditional expression of miRNA126 could modulate VEGF and BCL-2 expression in apoptosis-resistant endothelial cells. This lentiviral system specifically expressed miRNA126 in cells with high BCL-2 levels, downregulated VEGF expression, inhibited MAPK pathway activation and downregulated BCL-2 expression via suppression of AP1, and as a whole, reduced apoptosis-resistant endothelial cells, while the effects of miRNA126 on normal endothelial cells were relatively small. Our results demonstrate that conditional miRNA126 overexpression under the control of the downstream BCL-2 promoter provides a flexible regulatory strategy for reducing the apoptosis-resistant endothelial cells without having a significant impact on normal endothelial cells.
Collapse
Affiliation(s)
- Bo Liu
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - YiGang Li
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
50
|
Long ZJ, Wang LX, Zheng FM, Chen JJ, Luo Y, Tu XX, Lin DJ, Lu G, Liu Q. A novel compound against oncogenic Aurora kinase A overcomes imatinib resistance in chronic myeloid leukemia cells. Int J Oncol 2015; 46:2488-96. [PMID: 25872528 DOI: 10.3892/ijo.2015.2960] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 01/20/2015] [Indexed: 11/06/2022] Open
Abstract
Drug resistance still represents a major obstacle to successful chronic myeloid leukemia (CML) treatment and novel compounds or strategies to override this challenging problem are urgently required. Here, we evaluated a novel compound AKI603 against oncogenic Aurora kinase A (Aur-A) in imatinib-resistant CML cells. We found that Aur-A was highly activated in imatinib-resistant KBM5-T315I cells. AKI603 significantly inhibited the phosphorylation of Aur-A kinase at Thr288, while had little inhibitory effect on BCR-ABL kinase in both KBM5 and KBM5-T315I cells. AKI603 inhibited cell viability, and induced cell cycle arrest with polyploidy accumulation in KBM5 and KBM5-T315I cells. Moreover, inhibition of Aur-A kinase by AKI603 suppressed colony formation capacity without promoting obvious apoptosis. Importantly, AKI603 promoted cell differentiation in both CML cell types. Thus, our study suggested the potential clinical use of small molecule Aurora kinase inhibitor AKI603 to overcome imatinib resistance in CML treatment.
Collapse
Affiliation(s)
- Zi-Jie Long
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, Sun Yat-sen Institute of Hematology, Guangzhou 510630, P.R. China
| | - Le-Xun Wang
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, Sun Yat-sen Institute of Hematology, Guangzhou 510630, P.R. China
| | - Fei-Meng Zheng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Jia-Jie Chen
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, Sun Yat-sen Institute of Hematology, Guangzhou 510630, P.R. China
| | - Yu Luo
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P.R. China
| | - Xi-Xiang Tu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, P.R. China
| | - Dong-Jun Lin
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, Sun Yat-sen Institute of Hematology, Guangzhou 510630, P.R. China
| | - Gui Lu
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P.R. China
| | - Quentin Liu
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, Sun Yat-sen Institute of Hematology, Guangzhou 510630, P.R. China
| |
Collapse
|