1
|
Xie B, Dong L, Wang L, Wang R, Li C. Supramolecularly engineered bacteria mediated calcium overload and immunotherapy of tumors. Theranostics 2024; 14:6560-6570. [PMID: 39479452 PMCID: PMC11519789 DOI: 10.7150/thno.99931] [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: 06/21/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Intracellular Ca2+ nanogenerators, such as calcium carbonate, calcium peroxide, and calcium phosphate nanoparticles, have shown promise in calcium overload-mediated tumor therapy. However, their effectiveness is often hampered by poor targeting, low accumulation, and limited penetration into tumor cells, leading to suboptimal therapeutic outcomes. This strategy aims to achieve synergistic Ca2+ overload and immunotherapy of tumors. Methods: A supramolecular conjugate of engineered living bacteria (facultative anaerobic Salmonella typhimurium VNP20009, VNP) with CaCO3 nanoparticles was developed for targeted delivery of curcumin-loaded CaCO3 into tumors. Results: Both CaCO3 nanoparticles and the loaded Ca2+ efflux inhibiting agent, curcumin (CUR), demonstrated significant enhancement of intracellular Ca2+ overload, resulting in apoptosis of tumor cells via mitochondrial dysfunction. Moreover, VNP exhibited excellent tumor-targeting ability, colonization in tumor tissues, and anticancer activity with minimal side effects. Conclusion: The conjugate of VNP and CaCO3 not only enhances the efficiency of common cancer treatments but also synergizes Ca2+ overload with cancer immunotherapy, thereby offering a promising approach for improving therapeutic outcomes in cancer treatment.
Collapse
Affiliation(s)
- Beibei Xie
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai 200127, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau 999078, China
| | - Linmiao Dong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau 999078, China
| | - Leo Wang
- Kitsilano Secondary School, Vancouver, BC V6K 2J6, Canada
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau 999078, China
| | - Chunlai Li
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai 200127, China
| |
Collapse
|
2
|
Gao Y, Wang J, Zhang W, Ge F, Li W, Xu F, Cui T, Li X, Yang K, Tao Y. Application of capsaicin and calcium phosphate-loaded MOF system for tumor therapy involving calcium overload. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 60:102759. [PMID: 38851440 DOI: 10.1016/j.nano.2024.102759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/29/2024] [Accepted: 05/30/2024] [Indexed: 06/10/2024]
Abstract
Calcium overload therapy refers to the condition of intracellular Ca2+ overload, which causes mitochondrial damage and leads to the uncontrolled release of apoptotic factors into the cytoplasm through the open mitochondrial permeability pore. Based on this, it is playing an increasingly important role in the field of oncology due to its good efficacy and small side effects. However, the regulation of calcium homeostasis by cancer cells themselves, insufficient calcium ions (Ca2+) in tumor sites and low efficiency of calcium entering tumor have limited its efficacy, resulting in unsatisfactory therapeutic effect. Therefore, a novel CAP/BSA@TCP-ZIF-8 nanoparticle drug carrier system was constructed that can provide Ca2+ from exogenous sources for pH-controlled degradation and drug release at the same time. Both in vivo and in vitro experiments have proved that the nanomaterial can activate TRPV1 channels and provide exogenous Ca2+ to cause Ca2+ overload and apoptosis, thus achieving anti-tumor effects.
Collapse
Affiliation(s)
- Yuan Gao
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Jun Wang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Weiwei Zhang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Fei Ge
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Wanzhen Li
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Feiyang Xu
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Ting Cui
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Xing Li
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Yugui Tao
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China.
| |
Collapse
|
3
|
Chen X, Wei C, Zhao J, Zhou D, Wang Y, Zhang S, Zuo H, Dong J, Zhao Z, Hao M, He X, Bian Y. Carnosic acid: an effective phenolic diterpenoid for prevention and management of cancers via targeting multiple signaling pathways. Pharmacol Res 2024; 206:107288. [PMID: 38977208 DOI: 10.1016/j.phrs.2024.107288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/10/2024]
Abstract
Cancer is a serious global public health issue, and a great deal of research has been made to treat cancer. Of these, discovery of promising compounds that effectively fight cancer always has been the main point of interest in pharmaceutical research. Carnosic acid (CA) is a phenolic diterpenoid compound widely present in Lamiaceae plants such as Rosemary (Rosmarinus officinalis L.). In recent years, there has been increasing evidence that CA has significant anti-cancer activity, such as leukaemia, colorectal cancer, breast cancer, lung cancer, liver cancer, pancreatic cancer, stomach cancer, lymphoma, prostate cancer, oral cancer, etc. The potential mechanisms involved by CA, including inhibiting cell proliferation, inhibiting metastasis, inducing cell apoptosis, stimulating autophagy, regulating the immune system, reducing inflammation, regulating the gut microbiota, and enhancing the effects of other anti-cancer drugs. This article reviews the biosynthesis, pharmacokinetics and metabolism, safety and toxicity, as well as the molecular mechanisms and signaling pathways of the anticancer activity of CA. This will contribute to the development of CA or CA-containing functional foods for the prevention and treatment of cancer, providing important advances in the advancement of cancer treatment strategies.
Collapse
Affiliation(s)
- Xufei Chen
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, College of Life Science, Northwest University, Xi'an, Shaanxi 710069, China
| | - Cuntao Wei
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, College of Life Science, Northwest University, Xi'an, Shaanxi 710069, China
| | - Juanjuan Zhao
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, College of Life Science, Northwest University, Xi'an, Shaanxi 710069, China
| | - Dandan Zhou
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, College of Life Science, Northwest University, Xi'an, Shaanxi 710069, China
| | - Yue Wang
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, College of Life Science, Northwest University, Xi'an, Shaanxi 710069, China
| | - Shengxiang Zhang
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, College of Life Science, Northwest University, Xi'an, Shaanxi 710069, China
| | - Haiyue Zuo
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, College of Life Science, Northwest University, Xi'an, Shaanxi 710069, China
| | - Jianhui Dong
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, College of Life Science, Northwest University, Xi'an, Shaanxi 710069, China
| | - Zeyuan Zhao
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, College of Life Science, Northwest University, Xi'an, Shaanxi 710069, China
| | - Man Hao
- Clinical Medical College of Acuupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Department of Ortho and MSK Science, University College London, London WC1E 6BT, UK.
| | - Xirui He
- School of Bioengineering, Zhuhai Campus, Zunyi Medical University, Zhuhai, Guangdong 519041, China; UCL School of Pharmacy, Pharmacognosy & Phytotherapy, University College London, London WC1E 6BT, UK.
| | - Yangyang Bian
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, College of Life Science, Northwest University, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
4
|
Liu Z, Liu X, Zhang W, Gao R, Wei H, Yu CY. Current advances in modulating tumor hypoxia for enhanced therapeutic efficacy. Acta Biomater 2024; 176:1-27. [PMID: 38232912 DOI: 10.1016/j.actbio.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/08/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
Hypoxia is a common feature of most solid tumors, which promotes the proliferation, invasion, metastasis, and therapeutic resistance of tumors. Researchers have been developing advanced strategies and nanoplatforms to modulate tumor hypoxia to enhance therapeutic effects. A timely review of this rapidly developing research topic is therefore highly desirable. For this purpose, this review first introduces the impact of hypoxia on tumor development and therapeutic resistance in detail. Current developments in the construction of various nanoplatforms to enhance tumor treatment in response to hypoxia are also systematically summarized, including hypoxia-overcoming, hypoxia-exploiting, and hypoxia-disregarding strategies. We provide a detailed discussion of the rationale and research progress of these strategies. Through a review of current trends, it is hoped that this comprehensive overview can provide new prospects for clinical application in tumor treatment. STATEMENT OF SIGNIFICANCE: As a common feature of most solid tumors, hypoxia significantly promotes tumor progression. Advanced nanoplatforms have been developed to modulate tumor hypoxia to enhanced therapeutic effects. In this review, we first introduce the impact of hypoxia on tumor progression. Current developments in the construction of various nanoplatforms to enhance tumor treatment in response to hypoxia are systematically summarized, including hypoxia-overcoming, hypoxia-exploiting, and hypoxia-disregarding strategies. We discuss the rationale and research progress of the above strategies in detail, and finally introduce future challenges for treatment of hypoxic tumors. By reviewing the current trends, this comprehensive overview can provide new prospects for clinical translatable tumor therapy.
Collapse
Affiliation(s)
- Zihan Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xinping Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Wei Zhang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ruijie Gao
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
5
|
Yuan R, Zhang Y, Liao L, Ge Y, Li W, Zhi Q. Biomineralization-Inspired Anti-Caries Strategy Based on Multifunctional Nanogels as Mineral Feedstock Carriers. Int J Nanomedicine 2023; 18:4933-4947. [PMID: 37693886 PMCID: PMC10488770 DOI: 10.2147/ijn.s418465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/28/2023] [Indexed: 09/12/2023] Open
Abstract
Background Dentin caries remains a significant public concern, with no clinically viable material that effectively combines remineralization and antimicrobial properties. To address this issue, this study focused on the development of a bio-inspired multifunctional nanogel with both antibacterial and biomineralization properties. Methods First, p(NIPAm-co-DMC) (PNPDC) copolymers were synthesized from N-isopropylacrylamide (NIPAm) and 2-methacryloyloxyethyl-trimethyl ammonium chloride (DMC). Subsequently, PNPDC was combined with γ-polyglutamic acid (γ-PGA) through physical cross-linking to form nanogels. These nanogels served as templates for the mineralization of calcium phosphate (Cap), resulting in Cap-loaded PNPDC/PGA nanogels. The nanogels were characterized using various techniques, including TEM, particle tracking analysis, XRD, and FTIR. The release properties of ions were also assessed. In addition, the antibacterial properties of the Cap-loaded PNPDC/PGA nanogels were evaluated using the broth microdilution method and a biofilm formation assay. The remineralization effects were examined on both demineralized dentin and type I collagen in vitro. Results PNPDC/PGA nanogels were successfully synthesized and loaded with Cap. The diameter of the Cap-loaded PNPDC/PGA nanogels was measured as 196.5 nm at 25°C and 162.3 nm at 37°C. These Cap-loaded nanogels released Ca2+ and PO43- ions quickly, effectively blocking dental tubules with a depth of 10 μm and promoting the remineralization of demineralized dentin within 7 days. Additionally, they facilitated the heavy intrafibrillar mineralization of type I collagen within 3 days. Moreover, the Cap-loaded nanogels exhibited MIC50 and MIC90 values of 12.5 and 50 mg/mL against Streptococcus mutans, respectively, with an MBC value of 100 mg/mL. At a concentration of 50 mg/mL, the Cap-loaded nanogels also demonstrated potent inhibitory effects on biofilm formation by Streptococcus mutans while maintaining good biocompatibility. Conclusion Cap-loaded PNPDC/PGA nanogels are a multifunctional biomimetic system with antibacterial and dentin remineralization effects. This strategy of using antibacterial nanogels as mineral feedstock carriers offered fresh insight into the clinical management of caries.
Collapse
Affiliation(s)
- Rui Yuan
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510030, People’s Republic of China
| | - Yuwen Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510030, People’s Republic of China
| | - Liqiong Liao
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
| | - Yige Ge
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510030, People’s Republic of China
| | - Weichang Li
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510030, People’s Republic of China
| | - Qinghui Zhi
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510030, People’s Republic of China
| |
Collapse
|
6
|
Iweala EJ, Oluwapelumi AE, Dania OE, Ugbogu EA. Bioactive Phytoconstituents and Their Therapeutic Potentials in the Treatment of Haematological Cancers: A Review. Life (Basel) 2023; 13:1422. [PMID: 37511797 PMCID: PMC10381774 DOI: 10.3390/life13071422] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/12/2023] [Accepted: 02/15/2023] [Indexed: 07/30/2023] Open
Abstract
Haematological (blood) cancers are the cancers of the blood and lymphoid forming tissues which represents approximately 10% of all cancers. It has been reported that approximately 60% of all blood cancers are incurable. Despite substantial improvement in access to detection/diagnosis, chemotherapy and bone marrow transplantation, there is still high recurrence and unpredictable but clearly defined relapses indicating that effective therapies are still lacking. Over the past two decades, medicinal plants and their biologically active compounds are being used as potential remedies and alternative therapies for the treatment of cancer. This is due to their anti-oxidant, anti-inflammatory, anti-mutagenic, anti-angiogenic, anti-cancer activities and negligible side effects. These bioactive compounds have the capacity to reduce proliferation of haematological cancers via various mechanisms such as promoting apoptosis, transcription regulation, inhibition of signalling pathways, downregulating receptors and blocking cell cycle. This review study highlights the mechanistic and beneficial effects of nine bioactive compounds (quercetin, ursolic acid, fisetin, resveratrol, epigallocatechin gallate, curcumin, gambogic acid, butein and celastrol) as potential remedies for chemoprevention of haematological cancers. The study provides useful insights on the effectiveness of the use of bioactive compounds from plants for chemoprevention of haematological cancers.
Collapse
Affiliation(s)
- Emeka J Iweala
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota PMB 1023, Ogun State, Nigeria
- Covenant Applied Informatics and Communication African Centre of Excellence (CApIC-ACE), Covenant University, Ota PMB 1023, Ogun State, Nigeria
| | - Adurosakin E Oluwapelumi
- Department of Microbiology, Ladoke Akintola University of Technology, Ogbomoso PMB 4000, Oyo State, Nigeria
| | - Omoremime E Dania
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota PMB 1023, Ogun State, Nigeria
| | | |
Collapse
|
7
|
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: 6.0] [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
|
8
|
Chen L, Zhang X, Lin J, Wen Y, Chen Y, Chen CB. Construction and validation of a prognostic model based on stage-associated signature genes of head and neck squamous cell carcinoma: a bioinformatics study. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1316. [PMID: 36660709 PMCID: PMC9843360 DOI: 10.21037/atm-22-5427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022]
Abstract
Background Head and neck squamous cell carcinoma (HNSCC) is a malignancy of epithelial origin and with poor prognosis. Exploring the biomarkers and prognostic models that can contribute to early tumor detection is meaningful. A comprehensive analysis was conducted according to the stage-related signature genes of HNSCC, and a prognostic model was developed to validate their ability to predict the prognosis. Methods The transcriptome profiles and clinical information of HNSCC patients were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) respectively. mRNA expressions of differentially expressed genes (DEGs) were analyzed in stage I-II patients and stage III-IV patients from TCGA by R packages. A protein-protein interaction (PPI) network and core-gene network map were constructed, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to examine pathway enrichment. Kaplan-Meier, least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression were applied to establish a stage-associated signature model. A Spearman analysis was conducted to examine the correlations between the characteristic genes and immune cell infiltration. Kaplan-Meier analysis and a receiver operating characteristic (ROC) curve were used to test the effectiveness of the model. Univariate multivariate Cox regression analyses were used to assess whether the risk score was an independent prognostic indicator for HNSCC. Results In TCGA cohort, 5 genes (i.e., BRINP1, IL17A, ALB, FOXA2, and ZCCHC12) in the constructed prognostic risk model were associated with prognosis. Patients in the low-risk group had a better prognosis outcome than those in the high-risk group. The predictive power was good because all the area under the curve (AUC) of the risk score was higher than 0.6. Risk score [hazard ratio (HR) =1.985; P<0.001] was an independent risk factor for the prognosis of HNSCC. The results in the GEO cohort were consistent with those in the TCGA cohort. Conclusions We constructed and verified a prognostic risk model of stage-related signature genes for HNSCC based on the GEO and TCGA data. Due to the good predictive accuracy of this model, the prognosis of and the tumor immune cell infiltration with patients can be estimated.
Collapse
Affiliation(s)
- Lizhu Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China;,Cancer Bio-Immunotherapy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China;,Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, China
| | - Xiaofei Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China;,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jie Lin
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yaoming Wen
- Fujian Institute of Microbiology, Fuzhou, China
| | - Yu Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China;,Cancer Bio-Immunotherapy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China;,Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, China
| | - Chuan-Ben Chen
- Cancer Bio-Immunotherapy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China;,Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, China;,Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| |
Collapse
|
9
|
Acquaviva R, Malfa GA, Loizzo MR, Xiao J, Bianchi S, Tundis R. Advances on Natural Abietane, Labdane and Clerodane Diterpenes as Anti-Cancer Agents: Sources and Mechanisms of Action. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27154791. [PMID: 35897965 PMCID: PMC9330018 DOI: 10.3390/molecules27154791] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 01/14/2023]
Abstract
Extensive research over the past decades has identified numerous phytochemicals that could represent an important source of anti-cancer compounds. There is an immediate need for less toxic and more effective preventive and therapeutic strategies for the treatment of cancer. Natural compounds are considered suitable candidates for the development of new anti-cancer drugs due to their pleiotropic actions on target events with multiple manners. This comprehensive review highlighted the most relevant findings achieved in the screening of phytochemicals for anticancer drug development, particularly focused on a promising class of phytochemicals such as diterpenes with abietane, clerodane, and labdane skeleton. The chemical structure of these compounds, their main natural sources, and mechanisms of action were critically discussed.
Collapse
Affiliation(s)
- Rosaria Acquaviva
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria, 95125 Catania, Italy; (R.A.); (S.B.)
- CERNUT, Research Centre on Nutraceuticals and Health Products, Department of Drug and Health Sciences, University of Catania, Viale A. Doria, 95125 Catania, Italy
| | - Giuseppe A. Malfa
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria, 95125 Catania, Italy; (R.A.); (S.B.)
- CERNUT, Research Centre on Nutraceuticals and Health Products, Department of Drug and Health Sciences, University of Catania, Viale A. Doria, 95125 Catania, Italy
- Correspondence:
| | - Monica R. Loizzo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.R.L.); (R.T.)
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Sciences, Universidade de Vigo, 32004 Ourense, Spain;
| | - Simone Bianchi
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria, 95125 Catania, Italy; (R.A.); (S.B.)
| | - Rosa Tundis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.R.L.); (R.T.)
| |
Collapse
|
10
|
Hu T, Liu X, Gong X, Chen B, Tan S, Xu H, Pan A, Liang S, He Y, Zhou F. Multichannel Ca 2+ Generator for Synergistic Tumor Therapy via Intracellular Ca 2+ Overload and Chemotherapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:8012-8020. [PMID: 35715215 DOI: 10.1021/acs.langmuir.2c00794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Ca2+ overload has attracted an increasing attention due to its benefit of precise cancer therapy, but its efficacy is limited by the strong Ca2+ excretion of cancer cells. Moreover, monotherapy of Ca2+ overload usually fails to treat tumors satisfactorily. Herein, we develop a multifunctional nanosystem that could induce Ca2+ overload by multipathway and simultaneously produce chemotherapy for synergistic tumor therapy. The nanosystem (CaMSN@CUR) is prepared by synthesizing a Ca-doped mesoporous silica nanoparticle (CaMSN) followed by loading the anticancer drug curcumin (CUR). CaMSN serves as the basis Ca2+ generator to induce Ca2+ overload directly in the intracellular environment by acid-triggered Ca2+ release, while CUR could not only exhibit chemotherapy but also facilitate Ca2+ release from the endoplasmic reticulum to the cytoplasm and inhibit Ca2+ efflux out of cells to further enhance Ca2+ overload. The in vitro and in vivo results show that CaMSN@CUR could exhibit a remarkable cytotoxicity against 4T1 cells and significantly inhibit tumor growth in 4T1 tumor-bearing mice via the synergy of Ca2+ overload and CUR-mediated chemotherapy. It is expected that the designed CaMSN@CUR has a great potential for effective tumor therapy.
Collapse
Affiliation(s)
- Taishun Hu
- School of Materials Science and Engineering, Central South University, Changsha 410083, Hunan, China
| | - Xinli Liu
- School of Materials Science and Engineering, Central South University, Changsha 410083, Hunan, China
| | - Xiyu Gong
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Botao Chen
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan, China
| | - Songwen Tan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Hui Xu
- Institute of Super-Microstructure and Ultrafast Process in Advanced Materials, School of Physics and Electronics, Central South University, Changsha 410083, Hunan, China
| | - Anqiang Pan
- School of Materials Science and Engineering, Central South University, Changsha 410083, Hunan, China
| | - Shuquan Liang
- School of Materials Science and Engineering, Central South University, Changsha 410083, Hunan, China
| | - Yongju He
- School of Materials Science and Engineering, Central South University, Changsha 410083, Hunan, China
| | - Fangfang Zhou
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| |
Collapse
|
11
|
Cytotoxicity of Thioalkaloid-Enriched Nuphar lutea Extract and Purified 6,6′-Dihydroxythiobinupharidine in Acute Myeloid Leukemia Cells: The Role of Oxidative Stress and Intracellular Calcium. Pharmaceuticals (Basel) 2022; 15:ph15040410. [PMID: 35455407 PMCID: PMC9032197 DOI: 10.3390/ph15040410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/20/2022] [Accepted: 03/23/2022] [Indexed: 11/17/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematological malignancy characterized by uncontrolled proliferation of immature myeloid progenitors. Here, we report the in vitro antileukemic effects of the sesquiterpene thioalkaloid-enriched fraction of the Nuphar lutea leaf extract (NUP) and a purified thioalkaloid 6,6′-dihydroxythiobinupharidine (DTBN). Treatment with 0.3–10 µg/mL NUP caused a dose- and time-dependent reduction in proliferation and viability of human AML cells (KG-1a, HL60 and U937). This was associated with apoptosis induction manifested by annexin-V/propidium iodide binding as well as cleavage of caspases 8, 9, and 3 as well as poly (ADP-ribose) polymerase. Caspase-dependence of the apoptotic effect was confirmed using the pan-caspase inhibitor Q-VD-OPH. NUP induced significant biphasic changes in the cytosolic levels of reactive oxygen species (ROS) compared to untreated cells—a decrease at early time points (2–4 h) and an increase after a longer incubation (24 h). ROS accumulation was accompanied by lowering the cellular glutathione (GSH) levels. In addition, NUP treatment resulted in elevation of the cytosolic Ca2+ (Ca2+cyt) levels. The thiol antioxidant and glutathione precursor N-acetyl cysteine prevented NUP-induced ROS accumulation and markedly inhibited apoptosis. A similar antiapoptotic effect was obtained by Ca2+cyt chelating using BAPTA. These data indicate that NUP-induced cell death is mediated, at least in part, by the induction of oxidative stress and Ca2+cyt accumulation. However, a substantial apoptotic activity of pure DTBN (0.05–0.25 µg/mL), was found to be independent of cytosolic ROS or Ca2+, suggesting that alternative mechanisms are involved in DTBN-induced cytotoxicity. Notably, neither NUP nor DTBN treatment significantly induced cell death of normal human peripheral blood mononuclear cells. Our results provide the basis for further investigation of the antileukemic potential of NUP and its active constituents.
Collapse
|
12
|
Yang F, Wu A, Yao J, Peng H, Qiu Y, Li S, Xu X. Nanoplatform-mediated calcium overload for cancer therapy. J Mater Chem B 2022; 10:1508-1519. [DOI: 10.1039/d1tb02721b] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mitochondria, as the "the plant of power" of cells, have been extensively highlighted with biological functions of offering energy and participating in signaling pathways. In parallel, calcium (Ca2+) plays a...
Collapse
|
13
|
Braveboy-Wagner J, Sharoni Y, Lelkes PI. Nutraceuticals Synergistically Promote Osteogenesis in Cultured 7F2 Osteoblasts and Mitigate Inhibition of Differentiation and Maturation in Simulated Microgravity. Int J Mol Sci 2021; 23:136. [PMID: 35008559 PMCID: PMC8745420 DOI: 10.3390/ijms23010136] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/17/2021] [Accepted: 12/18/2021] [Indexed: 02/08/2023] Open
Abstract
Microgravity is known to impact bone health, similar to mechanical unloading on Earth. In the absence of countermeasures, bone formation and mineral deposition are strongly inhibited in Space. There is an unmet need to identify nutritional countermeasures. Curcumin and carnosic acid are phytonutrients with anticancer, anti-inflammatory, and antioxidative effects and may exhibit osteogenic properties. Zinc is a trace element essential for bone formation. We hypothesized that these nutraceuticals could counteract the microgravity-induced inhibition of osteogenic differentiation and function. To test this hypothesis, we cultured 7F2 murine osteoblasts in simulated microgravity (SMG) in a Random Positioning Machine in the presence and absence of curcumin, carnosic acid, and zinc and evaluated cell proliferation, function, and differentiation. SMG enhanced cell proliferation in osteogenic medium. The nutraceuticals partially reversed the inhibitory effects of SMG on alkaline phosphatase (ALP) activity and did not alter the SMG-induced reduction in the expression of osteogenic marker genes in osteogenic medium, while they promoted osteoblast proliferation and ALP activity in the absence of traditional osteogenic media. We further observed a synergistic effect of the intermix of the phytonutrients on ALP activity. Intermixes of phytonutrients may serve as convenient and effective nutritional countermeasures against bone loss in space.
Collapse
Affiliation(s)
- Justin Braveboy-Wagner
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA;
| | - Yoav Sharoni
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel;
| | - Peter I. Lelkes
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA;
| |
Collapse
|
14
|
Darawsha A, Trachtenberg A, Levy J, Sharoni Y. The Protective Effect of Carotenoids, Polyphenols, and Estradiol on Dermal Fibroblasts under Oxidative Stress. Antioxidants (Basel) 2021; 10:antiox10122023. [PMID: 34943127 PMCID: PMC8698602 DOI: 10.3390/antiox10122023] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/16/2021] [Accepted: 12/18/2021] [Indexed: 02/07/2023] Open
Abstract
Skin ageing is influenced by several factors including environmental exposure and hormonal changes. Reactive oxygen species (ROS), which mediate many of the effects of these factors, induce inflammatory processes in the skin and increase the production of matrix metalloproteinases (MMPs) in dermal fibroblasts, which leads to collagen degradation. Several studies have shown the protective role of estrogens and a diet rich in fruits and vegetables on skin physiology. Previous studies have shown that dietary carotenoids and polyphenols activate the cell’s antioxidant defense system by increasing antioxidant response element/Nrf2 (ARE/Nrf2) transcriptional activity and reducing the inflammatory response. The aim of the current study was to examine the protective effect of such dietary-derived compounds and estradiol on dermal fibroblasts under oxidative stress induced by H2O2. Human dermal fibroblasts were used to study the effect of H2O2 on cell number and apoptosis, MMP-1, and pro-collagen secretion as markers of skin damage. Treatment of cells with H2O2 led to cell death, increased secretion of MMP-1, and decreased pro-collagen secretion. Pre-treatment with tomato and rosemary extracts, and with estradiol, reversed the effects of the oxidative stress. This was associated with a reduction in intracellular ROS levels, probably through the measured increased activity of ARE/Nrf2. Conclusions: This study indicates that carotenoids, polyphenols, and estradiol protect dermal fibroblasts from oxidative stress-induced damage through a reduction in ROS levels.
Collapse
|
15
|
Structure-Activity Relationship of Hydroxycinnamic Acid Derivatives for Cooperating with Carnosic Acid and Calcitriol in Acute Myeloid Leukemia Cells. Biomedicines 2021; 9:biomedicines9111517. [PMID: 34829746 PMCID: PMC8615284 DOI: 10.3390/biomedicines9111517] [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] [Received: 07/15/2021] [Revised: 10/14/2021] [Accepted: 10/16/2021] [Indexed: 11/17/2022] Open
Abstract
Plant phenolic compounds have shown the ability to cooperate with one another at low doses in producing enhanced anticancer effects. This may overcome the limitations (e.g., poor bioavailability and high-dose toxicity) in developing these agents as cancer medicines. We have previously reported that the hydroxycinnamic acid derivative (HCAD) methyl-4-hydroxycinnamate and the phenolic diterpene carnosic acid (CA) can synergistically induce massive calcium-dependent apoptosis in acute myeloid leukemia (AML) at non-cytotoxic concentrations of each agent. Here, we explored the chemical nature of the synergy between HCADs and either CA, in inducing cytotoxicity, or the active metabolite of vitamin D (calcitriol), in enhancing the differentiation of AML cells. This was done by determining the structure–activity relationship of a series of hydroxycinnamic acids and their derivatives (methyl hydroxycinnamates and hydroxybenzylideneacetones) in combination with CA or calcitriol. The HCAD/CA synergy required the following critical structural elements of an HCAD molecule: (a) the para-hydroxyl on the phenolic ring, (b) the carbon C7–C8 double bond, and (c) the methyl-esterified carboxyl. Thus, the only HCADs capable of synergizing with CA were found to be methyl-4-hydroxycinnamate and methyl ferulate, which also most potently enhanced calcitriol-induced cell differentiation. Notably, the C7–C8 double bond was the major requirement for this HCAD/calcitriol cooperation. Our findings may contribute to the rational design of novel synergistically acting AML drugs based on prototype combinations of HCADs with other agents studied here.
Collapse
|
16
|
Curcumin and Carnosic Acid Cooperate to Inhibit Proliferation and Alter Mitochondrial Function of Metastatic Prostate Cancer Cells. Antioxidants (Basel) 2021; 10:antiox10101591. [PMID: 34679726 PMCID: PMC8533243 DOI: 10.3390/antiox10101591] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 12/24/2022] Open
Abstract
Anticancer activities of plant polyphenols have been demonstrated in various models of neoplasia. However, evidence obtained in numerous in vitro studies indicates that proliferation arrest and/or killing of cancer cells require quite high micromolar concentrations of polyphenols that are difficult to reach in vivo and can also be (geno)toxic to at least some types of normal cells. The ability of certain polyphenols to synergize with one another at low concentrations can be used as a promising strategy to effectively treat human malignancies. We have recently reported that curcumin and carnosic acid applied at non-cytotoxic concentrations synergistically cooperate to induce massive apoptosis in acute myeloid leukemia cells, but not in normal hematopoietic and non-hematopoietic cells, via sustained cytosolic calcium overload. Here, we show that the two polyphenols can also synergistically suppress the growth of DU145 and PC-3 metastatic prostate cancer cell cultures. However, instead of cell killing, the combined treatment induced a marked inhibition of cell proliferation associated with G0/G1 cell cycle arrest. This was preceded by transient elevation of cytosolic calcium levels and prolonged dissipation of the mitochondrial membrane potential, without generating oxidative stress, and was associated with defective oxidative phosphorylation encompassing mitochondrial dysfunction. The above effects were concomitant with a significant downregulation of mRNA and protein expression of the oncogenic kinase SGK1, the mitochondria-hosted mTOR component. In addition, a moderate decrease in SGK1 phosphorylation at Ser422 was observed in polyphenol-treated cells. The mTOR inhibitor rapamycin produced a similar reduction in SGK1 mRNA and protein levels as well as phosphorylation. Collectively, our findings suggest that the combination of curcumin and carnosic acid at potentially bioavailable concentrations may effectively target different types of cancer cells by distinct modes of action. This and similar combinations merit further exploration as an anticancer modality.
Collapse
|
17
|
Zoi V, Galani V, Lianos GD, Voulgaris S, Kyritsis AP, Alexiou GA. The Role of Curcumin in Cancer Treatment. Biomedicines 2021; 9:biomedicines9091086. [PMID: 34572272 PMCID: PMC8464730 DOI: 10.3390/biomedicines9091086] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [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
- Correspondence:
| |
Collapse
|
18
|
Zheng P, Ding B, Shi R, Jiang Z, Xu W, Li G, Ding J, Chen X. A Multichannel Ca 2+ Nanomodulator for Multilevel Mitochondrial Destruction-Mediated Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007426. [PMID: 33675268 DOI: 10.1002/adma.202007426] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/30/2020] [Indexed: 05/22/2023]
Abstract
Subcellular organelle-targeted nanoformulations for cancer theranostics are receiving increasing attention owing to their benefits of precise drug delivery, maximized therapeutic index, and reduced off-target side effects. Herein, a multichannel calcium ion (Ca2+ ) nanomodulator (CaNMCUR+CDDP ), i.e., a cisplatin (CDDP) and curcumin (CUR) co-incorporating calcium carbonate (CaCO3 ) nanoparticle, is prepared by a facile one-pot strategy in a sealed container with in situ synthesized polydopamine (PDA) as a template to enhance Ca2+ -overload-induced mitochondrial dysfunction in cancer therapy. After systemic administration, the PEGylated CaNMCUR+CDDP (PEG CaNMCUR+CDDP ) selectively accumulates in tumor tissues, enters tumor cells, and induces multilevel destruction of mitochondria by the combined effects of burst Ca2+ release, Ca2+ efflux inhibition by CUR, and chemotherapeutic CDDP, thereby observably boosting mitochondria-targeted tumor inhibition. Fluorescence imaging of CUR combined with photoacoustic imaging of PDA facilitates the visualization of the nanomodulator. The facile and practical design of this multichannel Ca2+ nanomodulator will contribute to the development of multimodal bioimaging-guided organelle-targeted cancer therapy in the future.
Collapse
Affiliation(s)
- Pan Zheng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, P. R. China
| | - Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Run Shi
- Faculty of Medicine, Ludwig-Maximilians-Universität München, Theresienstraße 39, D-80333, München, Germany
| | - Zhongyu Jiang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Weiguo Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Gao Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| |
Collapse
|
19
|
Alaswad HA, Mahbub AA, Le Maitre CL, Jordan-Mahy N. Molecular Action of Polyphenols in Leukaemia and Their Therapeutic Potential. Int J Mol Sci 2021; 22:ijms22063085. [PMID: 33802972 PMCID: PMC8002821 DOI: 10.3390/ijms22063085] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
Leukaemia is a malignant disease of the blood. Current treatments for leukaemia are associated with serious side-effects. Plant-derived polyphenols have been identified as potent anti-cancer agents and have been shown to work synergistically with standard chemotherapy agents in leukaemia cell lines. Polyphenols have multiple mechanisms of action and have been reported to decrease cell proliferation, arrest cell cycle and induce apoptosis via the activation of caspase (3, 8 and 9); the loss of mitochondrial membrane potential and the release of cytochrome c. Polyphenols have been shown to suppress activation of transcription factors, including NF-kB and STAT3. Furthermore, polyphenols have pro-oxidant properties, with increasing evidence that polyphenols inhibit the antioxidant activity of glutathione, causing oxidative DNA damage. Polyphenols also induce autophagy-driven cancer cell death and regulate multidrug resistance proteins, and thus may be able to reverse resistance to chemotherapy agents. This review examines the molecular mechanism of action of polyphenols and discusses their potential therapeutic targets. Here, we discuss the pharmacological properties of polyphenols, including their anti-inflammatory, antioxidant, anti-proliferative, and anti-tumour activities, and suggest that polyphenols are potent natural agents that can be useful therapeutically; and discuss why data on bioavailability, toxicity and metabolism are essential to evaluate their clinical use.
Collapse
Affiliation(s)
- Hamza A. Alaswad
- Biomolecular Sciences Research Centre, Department of Biosciences and Chemistry, Sheffield Hallam University, The Owen Building, City Campus, Howard Street, Sheffield S1 1WB, UK; (H.A.A.); (C.L.L.M.)
| | - Amani A. Mahbub
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia;
| | - Christine L. Le Maitre
- Biomolecular Sciences Research Centre, Department of Biosciences and Chemistry, Sheffield Hallam University, The Owen Building, City Campus, Howard Street, Sheffield S1 1WB, UK; (H.A.A.); (C.L.L.M.)
| | - Nicola Jordan-Mahy
- Biomolecular Sciences Research Centre, Department of Biosciences and Chemistry, Sheffield Hallam University, The Owen Building, City Campus, Howard Street, Sheffield S1 1WB, UK; (H.A.A.); (C.L.L.M.)
- Correspondence: ; Tel.: +44-0114-225-3120
| |
Collapse
|
20
|
Liu J, Zhu C, Xu L, Wang D, Liu W, Zhang K, Zhang Z, Shi J. Nanoenabled Intracellular Calcium Bursting for Safe and Efficient Reversal of Drug Resistance in Tumor Cells. NANO LETTERS 2020; 20:8102-8111. [PMID: 33064007 DOI: 10.1021/acs.nanolett.0c03042] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Multidrug resistance (MDR) of a tumor is the main cause of failure of clinical chemotherapy. Herein, we report a simple, yet versatile, tumor-targeting "calcium ion nanogenerator" (TCaNG) to reverse drug resistance by inducing intracellular Ca2+ bursting. Consequently, the TCaNG could induce Ca2+ bursting in acidic lysosomes of tumor cells and then reverse drug resistance according to the following mechanisms: (i) Ca2+ specifically accumulates in mitochondria, suppressing cellular respiration and relieving tumor hypoxia, thus inhibiting P-glycoprotein biosynthesis by downregulating HIF-1α expression. (ii) Ca2+-bursting-induced respiratory depression blocks intracellular ATP production, which further leads to the P-gp incompetence. As a result, the TCaNG could decrease the IC50 of DOX to MCF-7/ADR cells by approximately 30 times and reduce the proliferation of drug-resistant tumors by approximately 13 times without obvious side effects. This simple, safe, and effective "Ca2+ bursting" strategy holds the potential for clinical application in tumor treatment.
Collapse
Affiliation(s)
- Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, China, Zhengzhou University, Zhengzhou 450001, China
| | - Chunyu Zhu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Lihua Xu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Danyu Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wei Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, China, Zhengzhou University, Zhengzhou 450001, China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, China, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, China, Zhengzhou University, Zhengzhou 450001, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, China, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
21
|
Bahri S, Ali RB, Abdennabi R, Nahdi A, Mlika M, Jameleddine S. Industrial Elimination of Essential Oils from Rosmarinus Officinalis: In Support of the Synergic Antifibrotic Effect of Rosmarinic and Carnosic Acids in Bleomycin Model of Lung Fibrosis. Nutr Cancer 2020; 73:2376-2387. [PMID: 33059466 DOI: 10.1080/01635581.2020.1826991] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease characterized by collagen deposition as a consequence of excessive lung fibroblasts and myofibroblasts proliferation. We aimed to investigate for the first time the effect of rosemary leaf extract rich with carnosic acid (CA) or rosmarinic acid (RA), after industrial elimination of essential oils, against bleomycin (BLM)-induced lung fibrosis in rats. Male Wistar rats were given a single dose of BLM (4 mg/kg, intratracheal), while CA rich extract, RA rich extract or the combination RA/CA rich extracts (10, 75 and 150 mg/kg, intraperitoneal) were administered 3 day later and continued for 4 weeks. We reveled by HPLC an important similar amount of phenolic compounds such as pyrogallol, vanillic, gallic and ellagic acids in both rosemary extracts. BLM induced lung fibrotic foci and disturbance in superoxide dismutase, catalase and malondialdehyde levels. At 10 mg/kg, both rosemary extracts administrated alone or in combination alleviated synergistically lung fibrosis and ameliorated oxidative changes induced by BLM. In conclusion, industrial elimination of essential oils from rosemary allowed us to obtain two extracts with potent antifibrotic activities due to the large amount of RA and CA that appear much higher and effective than wild rosemary extract.
Collapse
Affiliation(s)
- Sana Bahri
- Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Laboratory of Physiopathology, Food and Biomolecules (LR-17-ES-03), Technology Center of Sidi Thabet, University of Manouba, Tunis, Tunisia.,Laboratory of Quality Control, Herbes De Tunisie, Company AYACHI-Group, Siliana, Tunisia
| | - Ridha Ben Ali
- Laboratory of Experimental Medicine, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Raed Abdennabi
- Laboratory of Plant Biotechnology, Faculty of Science, University of Sfax, Sfax, Tunisia
| | - Afef Nahdi
- Research Unit n° 17/ES/13, Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Mona Mlika
- Laboratory of Anatomy and Pathology, Abderhaman Mami Hospital, Ariana, Tunisia
| | - Saloua Jameleddine
- Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Laboratory of Physiopathology, Food and Biomolecules (LR-17-ES-03), Technology Center of Sidi Thabet, University of Manouba, Tunis, Tunisia
| |
Collapse
|
22
|
Dai Z, Tang J, Gu Z, Wang Y, Yang Y, Yang Y, Yu C. Eliciting Immunogenic Cell Death via a Unitized Nanoinducer. NANO LETTERS 2020; 20:6246-6254. [PMID: 32786942 DOI: 10.1021/acs.nanolett.0c00713] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Utilizing chemotherapeutics to induce immunogenic cell death (ICD) is a promising strategy to sensitize tumor cells and induce anticancer immunity. However, the application of traditional ICD inducers, such as chemodrugs, is largely hindered by their low tumor selectivity and severe side effects. Here, a new unitized ICD nanoinducer with high potency and cancer cell specificity is reported to achieve effective cancer immunotherapy. This nanoinducer is composed of disulfide-bond-incorporated organosilica nanoparticles, curcumin (CUR), and iron oxide nanoparticles, which can deplete intracellular glutathione, produce hydroxyl radicals, and induce cancer-cell-specific Ca2+ depletion as well as thioredoxin reductase inhibition. While the components are unable to induce ICD individually, their complementary pharmaceutical activities significantly elevate intracellular oxidative stress and endoplasmic reticulum stress in parallel. Consequently, ICD and systemic antitumor immunity can be elicited. Compared to the conventional ICD inducer doxorubicin, the unitized nanoinducer exhibits significantly improved ICD-inducing activity and cancer cell selectivity.
Collapse
Affiliation(s)
- Zan Dai
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Jie Tang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Zhengying Gu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Yue Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Yang Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Yannan Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, P.R. China
| |
Collapse
|
23
|
Carnosic acid increases sorafenib-induced inhibition of ERK1/2 and STAT3 signaling which contributes to reduced cell proliferation and survival of hepatocellular carcinoma cells. Oncotarget 2020; 11:3129-3143. [PMID: 32913557 PMCID: PMC7443370 DOI: 10.18632/oncotarget.27687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/07/2020] [Indexed: 11/25/2022] Open
Abstract
Hepatocellular carcinoma (HCC) has increasing worldwide incidence but when unresectable lacks curative options. Treatment with a kinase inhibitor Sorafenib (Sf), while initially effective, results in only short increases in patient survival, thus there is a need for improved treatment regimens. Numerous treatment regimens have been explored wherein Sf is combined with other agents, such as non-toxic botanicals like Curcumin or Silibinin. Recently, we have shown that carnosic acid (CA), a component of the food preservative Rosemary Extract, can markedly enhance the cytotoxic actions of Sf in several cell lines derived from HCC, but not in the cell line Hu1545 derived from normal hepatocytes. CA has been shown to enhance Sf-induced cell death in the neoplastic cell lines, principally due to the composite of increased apoptosis and cytotoxic autophagy. In the present study we focused on the mechanisms that underlie the reduced proliferation and survival of HCC cells when CA is added to Sf and how this relates to the increase in Sf-induced DNA damage as well as to the elevation of cytoplasmic levels of reactive oxygen species (ROS). Importantly, the elevation of ROS levels induced by Sf was increased by adding CA. We found that CA enhanced Sf-induced prolongation of cell cycle, and the overall decrease in cell growth was associated with reduced activation of both STAT3 transcription factor (TF) and extracellular signal-regulated protein kinase (Erk)1/2. Our data suggest that a regimen incorporating CA, an inexpensive and non-toxic food additive, in the treatment of advanced HCC merits clinical evaluation.
Collapse
|
24
|
Abstract
Ca2+ is a ubiquitous and dynamic second messenger molecule that is induced by many factors including receptor activation, environmental factors, and voltage, leading to pleiotropic effects on cell function including changes in migration, metabolism and transcription. As such, it is not surprising that aberrant regulation of Ca2+ signals can lead to pathological phenotypes, including cancer progression. However, given the highly context-specific nature of Ca2+-dependent changes in cell function, delineation of its role in cancer has been a challenge. Herein, we discuss the distinct roles of Ca2+ signaling within and between each type of cancer, including consideration of the potential of therapeutic strategies targeting these signaling pathways.
Collapse
Affiliation(s)
- Scott Gross
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Pranava Mallu
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Hinal Joshi
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Bryant Schultz
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Christina Go
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Jonathan Soboloff
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States; Department of Medical Genetics & Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.
| |
Collapse
|
25
|
Ma Z, Zhang J, Zhang W, Foda MF, Zhang Y, Ge L, Han H. Intracellular Ca 2+ Cascade Guided by NIR-II Photothermal Switch for Specific Tumor Therapy. iScience 2020; 23:101049. [PMID: 32334412 PMCID: PMC7183209 DOI: 10.1016/j.isci.2020.101049] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/16/2020] [Accepted: 04/05/2020] [Indexed: 10/29/2022] Open
Abstract
Currently, patients receiving cancer treatments routinely suffer from distressing toxic effects, most originating from premature drug leakage, poor biocompatibility, and off-targeting. For tackling this challenge, we construct an intracellular Ca2+ cascade for tumor therapy via photothermal activation of TRPV1 channels. The nanoplatform creates an artificial calcium overloading stress in specific tumor cells, which is responsible for efficient cell death. Notably, this efficient treatment is activated by mild acidity and TRPV1 channels simultaneously, which contributes to precise tumor therapy and is not limited to hypoxic tumor. In addition, Ca2+ possesses inherent unique biological effect and normal cells are more tolerant of the undesirable destructive influence than tumor cells. The Ca2+ overload leads to cell death due to mitochondrial dysfunction (upregulation of Caspase-3, cytochrome c, and downregulation of Bcl-2 and ATP), and in vivo, the released photothermal CuS nanoparticles allow an enhanced 3D photoacoustic imaging and provide instant diagnosis.
Collapse
Affiliation(s)
- Zhaoyu Ma
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, P. R. China
| | - Jin Zhang
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, P. R. China
| | - Weiyun Zhang
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, P. R. China
| | - Mohamed F Foda
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan, Hubei 430070, P. R. China; Department of Biochemistry Faculty of Agriculture, Benha University, Moshtohor, Toukh 13736, Egypt
| | - Yifan Zhang
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan, Hubei 430070, P. R. China
| | - Lin Ge
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan, Hubei 430070, P. R. China
| | - Heyou Han
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, P. R. China; State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan, Hubei 430070, P. R. China.
| |
Collapse
|
26
|
Klotho rewires cellular metabolism of breast cancer cells through alteration of calcium shuttling and mitochondrial activity. Oncogene 2020; 39:4636-4649. [PMID: 32398866 DOI: 10.1038/s41388-020-1313-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 02/07/2023]
Abstract
Klotho is a transmembrane protein, which can be shed and act as a circulating hormone and is involved in regulating cellular calcium levels and inhibition of the PI3K/AKT pathway. As a longevity hormone, it protects normal cells from oxidative stress, and as a tumor suppressor it inhibits growth of cancer cells. Mechanisms governing these differential activities have not been addressed. Altered cellular metabolism is a hallmark of cancer and dysregulation of mitochondrial activity is a hallmark of aging. We hypothesized that klotho exerts its differential effects through regulation of these two hallmarks. Treatment with klotho inhibited glycolysis, reduced mitochondrial activity and membrane potential only in cancer cells. Accordingly, global metabolic screen revealed that klotho altered pivotal metabolic pathways, amongst them glycolysis and tricarboxylic acid cycle in breast cancer cells. Alteration of metabolic activity and increased AMP/ATP ratio lead to LKB1-dependent AMPK activation. Indeed, klotho induced AMPK phosphorylation; furthermore, inhibition of LKB1 partially abolished klotho's tumor suppressor activity. By diminishing deltapsi (Δψ) klotho also inhibited mitochondria Ca2+ shuttling thereby impairing mitochondria communication with SOCE leading to reduced Ca2+ influx by SOCE channels. The reduced SOCE was followed by ER Ca2+ depletion and stress. These data delineate mechanisms mediating the differential effects of klotho toward cancer versus normal cells, and indicate klotho as a potent regulator of metabolic activity.
Collapse
|
27
|
Zhumina AG, Li K, Konovalova AA, Li YA, Ishmuratova MY, Pogossyan GP, Danilenko M. Plasma 25-Hydroxyvitamin D Levels and VDR Gene Expression in Peripheral Blood Mononuclear Cells of Leukemia Patients and Healthy Subjects in Central Kazakhstan. Nutrients 2020; 12:nu12051229. [PMID: 32357551 PMCID: PMC7281978 DOI: 10.3390/nu12051229] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/10/2020] [Accepted: 04/18/2020] [Indexed: 11/19/2022] Open
Abstract
Low blood levels of the vitamin D metabolite 25-hydroxyvitamin D [25(OH)D] have been associated with an increased risk and poorer outcomes of various cancers, including hematological malignancies. The Central Kazakhstan area has a relatively high incidence rate of leukemia. However, the relationship between vitamin D status and leukemia or other types of cancer in Kazakhstan has not yet been addressed. Therefore, in this first pilot single-center study conducted in Central Kazakhstan, we compared plasma levels of 25(OH)D and the vitamin D receptor (VDR) gene expression levels in peripheral blood mononuclear cells of patients with leukemia and demographically matching healthy volunteers. The levels of 25(OH)D in patients were found to be significantly lower (10.8 ± 7.0 ng/mL; n = 31) than in healthy subjects (21.6 ± 7.8 ng/mL; n = 34; p < 0.0001). A similar difference was observed in both younger (<60 years old) and older (>60 years old) participants, though there was no association between 25(OH)D concentration and age within the patient group. In female patients, 25(OH)D levels were significantly lower than in male patients (p = 0.04). No significant seasonal variations of 25(OH)D were observed in either the patient or the control group. VDR gene expression levels appeared to be similar in leukemia patients and healthy subjects, and no correlation between the cellular VDR expression and plasma 25(OH)D concentrations was observed in either group of participants. We did not observe a significant association of 25(OH)D or VDR levels and overall survival of leukemia patients. This observational study conducted for the first time in Kazakhstan supports previous findings demonstrating reduced blood 25(OH)D levels in cancer (leukemia) patients. Larger studies are required to determine whether low 25(OH)D plasma concentrations represent a risk factor for leukemia development and/or progression.
Collapse
Affiliation(s)
- Assel G. Zhumina
- Department of Botany, Academician Y.A. Buketov Karaganda State University, Karaganda 100028, Kazakhstan; (A.G.Z.); (A.A.K.); (M.Y.I.); (G.P.P.)
| | - Konstantin Li
- DNA Diagnostics Laboratory, the Dippner Health Center, Karaganda 100009, Kazakhstan; (K.L.); (Y.A.L.)
| | - Anna A. Konovalova
- Department of Botany, Academician Y.A. Buketov Karaganda State University, Karaganda 100028, Kazakhstan; (A.G.Z.); (A.A.K.); (M.Y.I.); (G.P.P.)
- DNA Diagnostics Laboratory, the Dippner Health Center, Karaganda 100009, Kazakhstan; (K.L.); (Y.A.L.)
| | - Yelena A. Li
- DNA Diagnostics Laboratory, the Dippner Health Center, Karaganda 100009, Kazakhstan; (K.L.); (Y.A.L.)
| | - Margarita Yu. Ishmuratova
- Department of Botany, Academician Y.A. Buketov Karaganda State University, Karaganda 100028, Kazakhstan; (A.G.Z.); (A.A.K.); (M.Y.I.); (G.P.P.)
| | - Gayane P. Pogossyan
- Department of Botany, Academician Y.A. Buketov Karaganda State University, Karaganda 100028, Kazakhstan; (A.G.Z.); (A.A.K.); (M.Y.I.); (G.P.P.)
- DNA Diagnostics Laboratory, the Dippner Health Center, Karaganda 100009, Kazakhstan; (K.L.); (Y.A.L.)
| | - Michael Danilenko
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- Correspondence: ; Tel.: +972-8647-9969
| |
Collapse
|
28
|
Wang C, Yu F, Liu X, Chen S, Wu R, Zhao R, Hu F, Yuan H. Cancer-Specific Therapy by Artificial Modulation of Intracellular Calcium Concentration. Adv Healthc Mater 2019; 8:e1900501. [PMID: 31368208 DOI: 10.1002/adhm.201900501] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/18/2019] [Indexed: 01/16/2023]
Abstract
Calcium (Ca2+ ) hemeostasis is crucial for the normal function of cellular biochemistry. The abnormal frequency of Ca2+ signaling in cancer cells makes them more vulnerable to Ca2+ modulation than normal cells. Here in this study, a novel cancer-specific therapy by artificially triggering Ca2+ overload in cancer cells is proposed. The feasibility of this therapy is illustrated by successful coupling of selective extrusion (Ca2+ ) inhibition effect of Curcumin (Cur) and the effective Ca2+ generating capability of amorphous calcium carbonate (ACC) into a facilely prepared water responsive phospholipid (PL)-ACC hybrid platform (PL/ACC-Cur). The obtained results demonstrate that PL/ACC-Cur can specifically boost the intracellular Ca2+ concentration to cause Ca2+ overload and to trigger mitochondria-related apoptosis in MCF-7 cells while sparing normal hepatocyte (L02), which might be a promising approach for effective cancer therapy.
Collapse
Affiliation(s)
- Cheng Wang
- College of Pharmaceutical SciencesZhejiang University 866 Yuhangtang Road Hangzhou 310058 China
- School of Food Science and Pharmaceutical EngineeringNanjing Normal University No. 1 Wenyuan Road Nanjing 210046 China
| | - Fangying Yu
- College of Pharmaceutical SciencesZhejiang University 866 Yuhangtang Road Hangzhou 310058 China
| | - Xuerong Liu
- College of Pharmaceutical SciencesZhejiang University 866 Yuhangtang Road Hangzhou 310058 China
| | - Shaoqing Chen
- College of Pharmaceutical SciencesZhejiang University 866 Yuhangtang Road Hangzhou 310058 China
| | - Rui Wu
- College of Pharmaceutical SciencesZhejiang University 866 Yuhangtang Road Hangzhou 310058 China
| | - Rui Zhao
- Sir Run Run Shaw HospitalSchool of MedicineZhejiang University No. 3 Qingchun East Road Hangzhou 310016 China
| | - Fuqiang Hu
- College of Pharmaceutical SciencesZhejiang University 866 Yuhangtang Road Hangzhou 310058 China
| | - Hong Yuan
- College of Pharmaceutical SciencesZhejiang University 866 Yuhangtang Road Hangzhou 310058 China
| |
Collapse
|
29
|
Nazıroğlu M, Çiğ B, Yazğan Y, Schwaerzer GK, Theilig F, Pecze L. Albumin evokes Ca 2+-induced cell oxidative stress and apoptosis through TRPM2 channel in renal collecting duct cells reduced by curcumin. Sci Rep 2019; 9:12403. [PMID: 31455864 PMCID: PMC6711968 DOI: 10.1038/s41598-019-48716-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 07/16/2019] [Indexed: 12/14/2022] Open
Abstract
In proteinuric nephropathies of chronic kidney disease, the epithelial cells of the nephron including the collecting duct are exposed to high concentrations of luminal albumin. Albumin is taken up from collecting duct cells by endocytosis causing excessive reactive oxygen species (ROS) production and a proinflammatory response. Curcumin used in the traditional medicine possesses anti-inflammatory and antioxidant effects. ROS and ADP-ribose (ADPR) activate the cation channel TRPM2. We hypothesize, that albumin-induced cell stress and proinflammatory response are mediated by Ca2+ and can be reduced by curcumin. The cortical collecting duct (CCD) cells mpkCCDc14 exhibit spontaneous and inducible Ca2+ oscillations, which can be blocked by pre-treatment with curcumin. Curcumin accumulates in plasma membrane and intracellular vesicles, where it interferes with TRPM2 and decreases the influx of Ca2+. Albumin reduces cell viability and increases apoptosis, NF-κB activation, and mitochondrial membrane depolarization via Ca2+-dependent signaling, which results in increased ROS production. Albumin-induced cell stress is diminished by the inhibition of TRPM2 after administration of curcumin and ADPR (PARP1) inhibitors. Curcumin did not reduce the Ca2+ elevation induced by thapsigargin in Ca2+-free medium, but it reduced the function of store-operated Ca2+ channels and ATP-evoked Ca2+ response. In conclusion, albumin-induced oxidative stress is mediated by Ca2+-dependent signaling via TRPM2 and leads to cell damage and a proinflammatory response, strengthening the role of CCD cells in the progression of chronic kidney disease.
Collapse
Affiliation(s)
- Mustafa Nazıroğlu
- Neuroscience Research Center, Suleyman Demirel University, Isparta, Turkey. .,Department of Biophysics, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey. .,Department of Neuroscience, Health Science Institute, Suleyman Demirel University, Isparta, Turkey.
| | - Bilal Çiğ
- Department of Biophysics, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey.,Department of Neuroscience, Health Science Institute, Suleyman Demirel University, Isparta, Turkey
| | - Yener Yazğan
- Department of Biophysics, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey.,Department of Neuroscience, Health Science Institute, Suleyman Demirel University, Isparta, Turkey
| | | | - Franziska Theilig
- Institute of Anatomy, Christian-Albrechts-University of Kiel, Kiel, Germany. .,Anatomy, Department of Medicine, University of Fribourg, Fribourg, Switzerland.
| | - László Pecze
- Anatomy, Department of Medicine, University of Fribourg, Fribourg, Switzerland.,Independent Scientist, Neuchhatel, Switzerland
| |
Collapse
|
30
|
Xiaoyu M, Xiuling D, Chunyu Z, Yi S, Jiangchao Q, Yuan Y, Changsheng L. Polyglutamic acid-coordinated assembly of hydroxyapatite nanoparticles for synergistic tumor-specific therapy. NANOSCALE 2019; 11:15312-15325. [PMID: 31386744 DOI: 10.1039/c9nr03176f] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Nanotechnology offers exciting and innovative therapeutic strategies in the fight against cancer. Nano-scale hydroxyapatite, the inorganic constituent of the hard tissues of humans and animals, is not only an ideal carrier for the delivery of drugs but also exerts selective inhibitory effects on tumor cells. To perform the dual functions, we propose polyglutamic acid-coordinated hydroxyapatite nanoparticles (HA-PGA NP) as both DOX delivery vehicle and sustained calcium flow supplier to achieve a synergistic, tumor-specific therapy in this study. With PGA as the coordinator, the HA-PGA NPs were easily assembled into spherical nano-clusters with low crystallinity. The excellent dispersibility and solubility in the tumor environment endowed the HA-PGA NPs with an improved internalization into the tumor cells, thereby causing a dramatic elevation in the intracellular calcium influx by about 40%, which further induced a cascade of mitochondrial membrane damage, ATP content reduction, and reinforced sensitivity to chemotherapy. After the encapsulation of the model drug DOX, a pH-responsive release profile was achieved via the degradation of the nanoparticles and the deprotonation of PGA in the acidic tumor micro-environment. Consequently, the hybrid system, with the synergistic effects of sustained DOX and calcium overload, exhibited selectively intensified toxicity to tumor cells. The in vivo test further confirmed that the current system exhibited highly selective tumor inhibition and reduced heart toxicity, thus representing an effective anti-tumor platform.
Collapse
Affiliation(s)
- Ma Xiaoyu
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China.
| | | | | | | | | | | | | |
Collapse
|
31
|
Koutsoulas A, Čarnecká M, Slanina J, Tóth J, Slaninová I. Characterization of Phenolic Compounds and Antiproliferative Effects of Salvia pomifera and Salvia fruticosa Extracts. Molecules 2019; 24:molecules24162921. [PMID: 31408993 PMCID: PMC6720736 DOI: 10.3390/molecules24162921] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/06/2019] [Accepted: 08/09/2019] [Indexed: 12/22/2022] Open
Abstract
The phenolic compounds of methanolic extracts of Salvia pomifera and Salvia fruticosa were identified by liquid chromatography tandem mass spectrometry. Carnosic acid and its metabolite carnosol were the most abundant terpene phenolic compounds of S. fruticosa, while they were completely absent in S. pomifera. The main terpene phenolic constituent of S. pomifera was 12-O-methylcarnosic acid and its mass/mass fragmentation pathway was explained. The detailed mechanism of carnosic acid oxidation to carnosol was suggested. The effects of Salvia extracts and/or carnosic acid, the main diterpene phenolic component of S. fruticosa, on the proliferation and cell cycle of two melanoma cell lines (A375, Mel JuSo) and human fibroblast cell line (HFF) were investigated by MTT assay, PI-exclusion assay and flow cytometry cell cycle analysis. Extract of S. fruticosa more efficiently than S. pomifera extract reduced the proliferation of the human melanoma cells. Carnosic acid showed the most significant effect. The first evidence that carnosic acid affects microtubule dynamics and arrests the cell cycle in the G2/M phase was provided. Collectively, our results demonstrate that these two Salvia species are plants of medicinal interest with perspective for further investigation. Carnosic acid could be the compound responsible for the biological activities of S. fruticosa extracts.
Collapse
Affiliation(s)
- Antonios Koutsoulas
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 83232 Bratislava 3, Slovak Republic
| | - Martina Čarnecká
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Kamenice 5, Building A16, 62500 Brno, Czech Republic
| | - Jiří Slanina
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Kamenice 5, Building A16, 62500 Brno, Czech Republic
| | - Jaroslav Tóth
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 83232 Bratislava 3, Slovak Republic.
| | - Iva Slaninová
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5, Building A6, 62500 Brno, Czech Republic.
| |
Collapse
|
32
|
Trachtenberg A, Muduli S, Sidoryk K, Cybulski M, Danilenko M. Synergistic Cytotoxicity of Methyl 4-Hydroxycinnamate and Carnosic Acid to Acute Myeloid Leukemia Cells via Calcium-Dependent Apoptosis Induction. Front Pharmacol 2019; 10:507. [PMID: 31143124 PMCID: PMC6521573 DOI: 10.3389/fphar.2019.00507] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/24/2019] [Indexed: 01/15/2023] Open
Abstract
Acute myeloid leukemia (AML) is a malignant hematopoietic disease with poor prognosis for most patients. Conventional chemotherapy has been the standard treatment approach for AML in the past 40 years with limited success. Although, several targeted drugs were recently approved, their long-term impact on survival of patients with AML is yet to be determined. Thus, it is still necessary to develop alternative therapeutic approaches for this disease. We have previously shown a marked synergistic anti-leukemic effect of two polyphenols, curcumin (CUR) and carnosic acid (CA), on AML cells in-vitro and in-vivo. In this study, we identified another phenolic compound, methyl 4-hydroxycinnamate (MHC), which among several tested phytochemicals could uniquely cooperate with CA in killing AML cells, but not normal peripheral blood mononuclear cells. Notably, our data revealed striking phenotypical and mechanistic similarities in the apoptotic effects of MHC+CA and CUR+CA on AML cells. Yet, we show that MHC is a non-fluorescent molecule, which is an important technical advantage over CUR that can interfere in various fluorescence-based assays. Collectively, we demonstrated for the first time the antileukemic activity of MHC in combination with another phenolic compound. This type of synergistically acting combinations may represent prototypes for novel antileukemic therapy.
Collapse
Affiliation(s)
- Aviram Trachtenberg
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Suchismita Muduli
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Katarzyna Sidoryk
- Chemistry Department, Pharmaceutical Research Institute, Warsaw, Poland
| | - Marcin Cybulski
- Chemistry Department, Pharmaceutical Research Institute, Warsaw, Poland
| | - Michael Danilenko
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
33
|
Nachliely M, Trachtenberg A, Khalfin B, Nalbandyan K, Cohen-Lahav M, Yasuda K, Sakaki T, Kutner A, Danilenko M. Dimethyl fumarate and vitamin D derivatives cooperatively enhance VDR and Nrf2 signaling in differentiating AML cells in vitro and inhibit leukemia progression in a xenograft mouse model. J Steroid Biochem Mol Biol 2019; 188:8-16. [PMID: 30508646 DOI: 10.1016/j.jsbmb.2018.11.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/12/2018] [Accepted: 11/29/2018] [Indexed: 12/11/2022]
Abstract
Acute myeloid leukemia (AML) is one of the deadliest hematological malignancies without effective treatment for most patients. Vitamin D derivatives (VDDs) - active metabolites 1α,25-dihydroxyvitamin D2 (1,25D2) and 1α,25-dihydroxyvitamin D3 (1,25D3) and their analogs - are differentiation-inducing agents which have potential for the therapy of AML. However, calcemic toxicity of VDDs limits their clinical use at doses effective against cancer cells in vivo. Here, we demonstrate that in AML cell cultures, moderate pro-differentiation effects of low concentrations of VDDs can be synergistically enhanced by structurally distinct compounds known to activate the transcription factor Nuclear Factor (Erythroid-derived 2)-Like 2 (NFE2L2 or Nrf2). Particularly, dimethyl fumarate (DMF), which is clinically approved for the treatment of multiple sclerosis and psoriasis, strongly cooperated with 1,25D3, PRI-5100 (19-nor-1,25D2; paricalcitol) and PRI-5202 (a double-point modified 19-nor analog of 1,25D2). The pro-differentiation synergy between VDDs (1,25D3 or PRI-5202) and Nrf2 activators (DMF, tert-butylhydroquinone or carnosic acid) was associated with a cooperative upregulation of the protein levels of the vitamin D receptor (VDR) and Nrf2 as well as increased mRNA expression of their respective target genes. These data support the notion that VDDs and Nrf2 activators synergize in inducing myeloid cell differentiation through the cooperative activation of the VDR and Nrf2/antioxidant response element signaling pathways. We have previously reported that PRI-5202 is more potent by approximately two orders of magnitude than 1,25D3 as a differentiation inducer in AML cell lines. In this study, we found that PRI-5202 was also at least 5-fold less calcemic in healthy mice compared to both its direct precursor PRI-1907 and 1,25D3. In addition, PRI-5202 was remarkably more resistant against degradation by the human 25-hydroxyvitamin D3-24-hydroxylase than both 1,25D2 and 1,25D3. Importantly, using a xenograft mouse model we demonstrated that co-administration of PRI-5202 and DMF resulted in a marked cooperative inhibition of human AML tumor growth without inducing treatment toxicity. Collectively, our findings provide a rationale for clinical testing of low-toxic VDD/DMF combinations as a novel approach for differentiation therapy of AML.
Collapse
Affiliation(s)
- Matan Nachliely
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105 Beer Sheva, Israel
| | - Aviram Trachtenberg
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105 Beer Sheva, Israel
| | - Boris Khalfin
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105 Beer Sheva, Israel
| | - Karen Nalbandyan
- Department of Pathology, Soroka University Medical Center, and Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105 Beer-Sheva, Israel
| | - Merav Cohen-Lahav
- Laboratory of Biochemistry, Soroka University Medical Center, 84101 Beer Sheva, Beer Sheva, Israel
| | - Kaori Yasuda
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, 939-0398 Imizu, Toyama, Japan
| | - Toshiyuki Sakaki
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, 939-0398 Imizu, Toyama, Japan
| | - Andrzej Kutner
- Department of Pharmacology, Pharmaceutical Research Institute, 01-793 Warsaw, Poland
| | - Michael Danilenko
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105 Beer Sheva, Israel.
| |
Collapse
|
34
|
Wang X, Nachliely M, Harrison JS, Danilenko M, Studzinski GP. Participation of vitamin D-upregulated protein 1 (TXNIP)-ASK1-JNK1 signalosome in the enhancement of AML cell death by a post-cytotoxic differentiation regimen. J Steroid Biochem Mol Biol 2019; 187:166-173. [PMID: 30508644 PMCID: PMC6501208 DOI: 10.1016/j.jsbmb.2018.11.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/02/2018] [Accepted: 11/29/2018] [Indexed: 01/06/2023]
Abstract
Standard therapy for Acute Myeloid Leukemia (AML) is rarely curative, and several suggested improvements have had little success so far. We have reported that in an in vitro model of a potential therapeutic regimen for AML, the activity of cytarabine (AraC) is enhanced by a sequential treatment with a combination of the vitamin D2 analog Doxercalciferol (D2) and the plant-derived antioxidant carnosic acid (CA). Importantly, the enhancement occurred selectively in patient-derived AML blasts, but not in the normal bone marrow cells. We now demonstrate that TXNIP, previously known as Vitamin D up-regulated protein 1 (VDUP1) [PMID 808674] plays a part in signaling cell death (CD) in this regimen. This is shown by the reduced CD when TXNIP protein levels are decreased by the CRISPR/CAS9 or RNAi technology. Further, we show that direct activation of ASK1 kinase by TXNIP is required for the optimal transmission of the CD signal to apoptotic machinery, regulated by JNK and BIM. These studies provide a rationale for a projected clinical trial of this vitamin D-based new therapeutic regimen for AML.
Collapse
Affiliation(s)
- X Wang
- Department of Pathology & Laboratory Medicine, Rutgers NJ Medical School, Newark, NJ, United States
| | - M Nachliely
- Department of Clinical Biochemistry & Pharmacology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - J S Harrison
- University of Connecticut School of Medicine, Farmington, CT, United States
| | - M Danilenko
- Department of Clinical Biochemistry & Pharmacology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - G P Studzinski
- Department of Pathology & Laboratory Medicine, Rutgers NJ Medical School, Newark, NJ, United States.
| |
Collapse
|
35
|
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: 37] [Impact Index Per Article: 7.4] [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
|
36
|
The Cooperative Relationship between STAT5 and Reactive Oxygen Species in Leukemia: Mechanism and Therapeutic Potential. Cancers (Basel) 2018; 10:cancers10100359. [PMID: 30262727 PMCID: PMC6210354 DOI: 10.3390/cancers10100359] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/21/2018] [Accepted: 09/24/2018] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are now recognized as important second messengers with roles in many aspects of signaling during leukemogenesis. They serve as critical cell signaling molecules that regulate the activity of various enzymes including tyrosine phosphatases. ROS can induce inactivation of tyrosine phosphatases, which counteract the effects of tyrosine kinases. ROS increase phosphorylation of many proteins including signal transducer and activator of transcription-5 (STAT5) via Janus kinases (JAKs). STAT5 is aberrantly activated through phosphorylation in many types of cancer and this constitutive activation is associated with cell survival, proliferation, and self-renewal. Such leukemic activation of STAT5 is rarely caused by mutation of the STAT5 gene itself but instead by overactive mutant receptors with tyrosine kinase activity as well as JAK, SRC family protein tyrosine kinases (SFKs), and Abelson murine leukemia viral oncogene homolog (ABL) kinases. Interestingly, STAT5 suppresses transcription of several genes encoding antioxidant enzymes while simultaneously enhancing transcription of NADPH oxidase. By doing so, STAT5 activation promotes an overall elevation of ROS level, which acts as a feed-forward loop, especially in high risk Fms-related tyrosine kinase 3 (FLT3) mutant leukemia. Therefore, efforts have been made recently to target ROS in cancer cells. Drugs that are able to either quench ROS production or inversely augment ROS-related signaling pathways both have potential as cancer therapies and may afford some selectivity by activating feedback inhibition of the ROS-STAT5 kinome. This review summarizes the cooperative relationship between ROS and STAT5 and explores the pros and cons of emerging ROS-targeting therapies that are selective for leukemia characterized by persistent STAT5 phosphorylation.
Collapse
|
37
|
Xu L, Tong G, Song Q, Zhu C, Zhang H, Shi J, Zhang Z. Enhanced Intracellular Ca 2+ Nanogenerator for Tumor-Specific Synergistic Therapy via Disruption of Mitochondrial Ca 2+ Homeostasis and Photothermal Therapy. ACS NANO 2018; 12:6806-6818. [PMID: 29966081 DOI: 10.1021/acsnano.8b02034] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Breast cancer therapy has always been a hard but urgent issue. Disruption of mitochondrial Ca2+ homeostasis has been reported as an effective antitumor strategy, while how to contribute to mitochondrial Ca2+ overload effectively is a critical issue. To solve this issue, we designed and engineered a dual enhanced Ca2+ nanogenerator (DECaNG), which can induce elevation of intracellular Ca2+ through the following three ways: Calcium phosphate (CaP)-doped hollow mesoporous copper sulfide was the basic Ca2+ nanogenerator to generate Ca2+ directly and persistently in the lysosomes (low pH). Near-infrared light radiation (NIR, such as 808 nm laser) can accelerate Ca2+ generation from the basic Ca2+ nanogenerator by disturbing the crystal lattice of hollow mesoporous copper sulfide via NIR-induced heat. Curcumin can facilitate Ca2+ release from the endoplasmic reticulum to cytoplasm and inhibit expelling of Ca2+ in cytoplasm through the cytoplasmic membrane. The in vitro study showed that DECaNG could produce a large amount of Ca2+ directly and persistently to flow to mitochondria, leading to upregulation of Caspase-3, cytochrome c, and downregulation of Bcl-2 and ATP followed by cell apoptosis. In addition, DECaNG had an outstanding photothermal effect. Interestingly, it was found that DECaNG exerted a stronger photothermal effect at lower pH due to the super small nanoparticles effect, thus enhancing photothermal therapy. In the in vivo study, the nanoplatform had good tumor targeting and treatment efficacy via a combination of disruption of mitochondrial Ca2+ homeostasis and photothermal therapy. The metabolism of CaNG was sped up through disintegration of CaNG into smaller nanoparticles, reducing the retention time of the nanoplatform in vivo. Therefore, DECaNG can be a promising drug delivery system for breast cancer therapy.
Collapse
Affiliation(s)
- Lihua Xu
- School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou , People's Republic of China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province , People's Republic of China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province , Zhengzhou , China
| | - Guihua Tong
- School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou , People's Republic of China
| | - Qiaoli Song
- School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou , People's Republic of China
| | - Chunyu Zhu
- School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou , People's Republic of China
| | - Hongling Zhang
- School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou , People's Republic of China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province , People's Republic of China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province , Zhengzhou , China
| | - Jinjin Shi
- School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou , People's Republic of China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province , People's Republic of China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province , Zhengzhou , China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou , People's Republic of China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province , People's Republic of China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province , Zhengzhou , China
| |
Collapse
|
38
|
Garrido-Armas M, Corona JC, Escobar ML, Torres L, Ordóñez-Romero F, Hernández-Hernández A, Arenas-Huertero F. Paraptosis in human glioblastoma cell line induced by curcumin. Toxicol In Vitro 2018; 51:63-73. [PMID: 29723631 DOI: 10.1016/j.tiv.2018.04.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 04/27/2018] [Accepted: 04/28/2018] [Indexed: 12/20/2022]
Abstract
Curcumin is a polyphenol compound extracted from Curcuma longa plant, is a molecule with pleiotropic effects that suppresses transformation, proliferation and metastasis of malignant tumors. Curcumin can cause different kinds of cell death depending of its concentration on the exposed cell type. Here we show that exposure of the glioblastoma cell line A172 to curcumin at 50 μM, the IC50, causes morphological change characteristic of paraptosis cell-death. Vesicles derived from the endoplasmic reticulum (ER) and low membrane potential of the mitochondria were constantly found in the exposed cells. Furthermore, changes in expression of the ER Stress Response (ERSR) genes IRE1 and ATF6, and the microRNAs (miRNAs) miR-27a, miR-222, miR-449 was observed after exposure to curcumin. AKT-Insulin and p53-BCL2 networks were predicted being modulated by the affected miRNAs. Furthermore, AKT protein levels reduction was confirmed. Our data, strongly suggest that curcumin exerts its cell-death properties by affecting the integrity of the reticulum, leading to paraptosis in the glioblastoma cells. These data unveils the versatility of curcumin to control cancer progression.
Collapse
Affiliation(s)
| | - Juan Carlos Corona
- Laboratorio de Neurociencias, Hospital Infantil de México Federico Gómez, Av. Dr. Márquez 162. Colonia Doctores, C.P. 06720 Ciudad de México, Mexico
| | - Maria Luisa Escobar
- Departamento de Biología Celular, Laboratorio de Microscopía Electrónica, Facultad de Ciencias, Universidad Nacional Autónoma de México (UNAM), Av. Universidad 3000, Circuito Exterior S/N Delegación Coyoacán, C.P. 04510. Ciudad Universitaria, Ciudad de México, México
| | - Leda Torres
- Laboratorio de Citogenética, Departamento de Investigación en Genética Humana, Instituto Nacional de Pediatría, Av. Insurgentes Sur 3700, Letra C, Col. Insurgentes Cuicuilco, Delegación Coyoacán, C.P. 04530 Ciudad de México, México
| | | | | | | |
Collapse
|
39
|
Personalizing Chinese medicine by integrating molecular features of diseases and herb ingredient information: application to acute myeloid leukemia. Oncotarget 2018; 8:43579-43591. [PMID: 28454110 PMCID: PMC5522171 DOI: 10.18632/oncotarget.16983] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/06/2017] [Indexed: 11/30/2022] Open
Abstract
Traditional Chinese Medicine (TCM) has been widely used as a complementary medicine in Acute Myeloid Leukemia (AML) treatment. In this study, we proposed a new classification of Chinese Medicines (CMs) by integrating the latest discoveries in disease molecular mechanisms and traditional medicine theory. We screened out a set of chemical compounds on basis of AML differential expression genes and chemical-protein interactions and then mapped them to Traditional Chinese Medicine Integrated Database. 415 CMs contain those compounds and they were categorized into 8 groups according to the Traditional Chinese Pharmacology. Pathway analysis and synthetic lethality gene pairs were applied to analyze the dissimilarity, generality and intergroup relations of different groups. We defined hub CM pairs and alternative CM groups based on the analysis result and finally proposed a formula to form an effective anti-AML prescription which combined the hub CM pairs with alternative CMs according to patients’ molecular features. Our method of formulating CMs based on patients’ stratification provides novel insights into the new usage of conventional CMs and will promote TCM modernization.
Collapse
|
40
|
The c-Raf modulator RRD-251 enhances nuclear c-Raf/GSK-3/VDR axis signaling and augments 1,25-dihydroxyvitamin D3-induced differentiation of HL-60 myeloblastic leukemia cells. Oncotarget 2018. [PMID: 29515772 PMCID: PMC5839403 DOI: 10.18632/oncotarget.24275] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Differentiation therapy is used in cancer treatment. Epidemiologic studies showed that higher vitamin D levels are associated with reduced cancer risks. However, the therapeutic doses needed for differentiation are accompanied by hypercalcemia and intolerable pathological sequelae. In the present work we evaluated if RRD-251, a small-molecule, can enhance vitamin D3-induced differentiation of leukemic cells, in the hope of decreasing the needed vitamin D3-dose. We demonstrate that RRD-251 enhances vitamin D3-induced differentiation of leukemic cells, the enrichment of the c-Raf kinase in the nucleus, the binding of nuclear c-Raf to GSK-3, increased phosphorylation of GSK-3 ser 21/9 inhibitory sites, and the binding of GSK-3 to VDR, where GSK-3 inhibition is known to enhance transcriptional activation by VDR. Enhancement of D3-induced p-GSK-3 ser 21/9 by RRD-251 was associated with enhanced Akt-GSK-3 binding, Akt being a known GSK-3 inhibitor, and diminished Erk1/2 binding. Diminishing Erk interaction with GSK-3 was associated with enhanced interaction with Vav1, a known driver of myeloid differentiation. This is redolent of an ATRA/c-Raf/GSK-3/RARα axis we previously reported, although the phosphorylation effects to enhance transcriptional activation on RARα vs VDR diverge. Taken together this indicates potential therapeutic significance for a c-Raf/GSK-3/VDR or RARα axis for leukemic myelo-monocytic differentiation.
Collapse
|
41
|
Bahri S, Ben Ali R, Gasmi K, Mlika M, Fazaa S, Ksouri R, Serairi R, Jameleddine S, Shlyonsky V. Prophylactic and curative effect of rosemary leaves extract in a bleomycin model of pulmonary fibrosis. PHARMACEUTICAL BIOLOGY 2017; 55:462-471. [PMID: 28093019 PMCID: PMC6130597 DOI: 10.1080/13880209.2016.1247881] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 09/22/2016] [Accepted: 10/09/2016] [Indexed: 06/01/2023]
Abstract
CONTEXT Pulmonary fibrosis is a devastating disease without effective treatment. Rosemary is appreciated since ancient times for its medicinal properties, while biomolecules originated from the plant have an antioxidant and antifibrotic effect. OBJECTIVE The effects of Rosmarinus officinalis L. (Lamiaceae) leaves extract (RO) on bleomycin-induced lung fibrosis were investigated. MATERIALS AND METHODS Male Wistar rats were given a single dose of bleomycin (BLM, 4 mg/kg, intratracheal), while RO (75 mg/kg, intraperitoneal) was administered 3 days later and continued for 4 weeks (BLM/RO1-curative group). Alternatively, RO was administered 2 weeks before BLM and continued 15 days thereafter (BLM/RO2-prophylactic group). Antioxidant activities of RO and lung tissues were studied by standard methods. Histological staining revealed lung architecture and collagen deposition. RO was characterized for its polyphenol content and by high-performance liquid chromatography. RESULTS RO polyphenol content was 60.52 mg/g of dry weight, carnosic and rosmarinic acids being major components (6.886 and 2.351 mg/g). Antioxidant effect of RO (DPPH and FRAP assay) expressed as IC50 values were 2.23 μg/mL and 0.074 μg/mL, respectively. In BLM/RO1 and BLM/RO2 lung architecture was less compromised compared to BLM, which was reflected in lower fibrosis score (2.33 ± 0.33 and 1.8 ± 0.32 vs 3.7 ± 0.3). Malondialdehyde levels were attenuated (141% and 108% vs 258% of normal value). Catalase and glutathione-S-transferase activities were normalized (103% and 117% vs 59%, 85% and 69% vs 23%, respectively). DISCUSSION AND CONCLUSION RO has a protective effect against BLM-induced oxidative stress and lung fibrosis due to its phenolic compounds.
Collapse
Affiliation(s)
- Sana Bahri
- Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Physiopathology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Ridha Ben Ali
- Laboratory of Experimental Medicine, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Khaoula Gasmi
- Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Mona Mlika
- Laboratory of Anatomy and Pathology, Abderhaman Mami Hospital, Ariana, Tunisia
| | - Saloua Fazaa
- Laboratory of Physiology, Faculty of Science of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Riadh Ksouri
- Laboratory of Eco-Process and Valorization of Aromatic and Medicinal Plants, Center for Biotechnology, Technopole Borj Cédria (CBBC), Tunis, Tunisia
| | - Raja Serairi
- Laboratory of Physiology, Faculty of Science of Tunis, University of Tunis El Manar, Tunis, Tunisia
- High School of Health Sciences, Tunis, Tunisia
| | - Saloua Jameleddine
- Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Vadim Shlyonsky
- Laboratory of Physiopathology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
42
|
Aralbaeva AN, Mamataeva AT, Zhaparkulova NI, Utegalieva RS, Khanin M, Danilenko M, Murzakhmetova MK. A composition of medicinal plants with an enhanced ability to suppress microsomal lipid peroxidation and a protective activity against carbon tetrachloride-induced hepatotoxicity. Biomed Pharmacother 2017; 96:1283-1291. [PMID: 29169731 DOI: 10.1016/j.biopha.2017.11.085] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 11/02/2017] [Accepted: 11/16/2017] [Indexed: 01/05/2023] Open
Abstract
Treatment of liver injury induced by various toxicants represents a serious clinical challenge. Here, we utilized the ability of natural agents to inhibit microsomal lipid peroxidation (LPO) as the in-vitro screening paradigm for selecting efficacious tissue-protective combinations of cooperatively acting medicinal plants. Based on screening of 70 water-ethanol extracts obtained from different parts of 65 plants we prepared a highly active phytocomposition (PC-1) containing oregano (Origanum vulgare), wild thyme (Thymus serpyllum) and coltsfoot (Tussilago farfara) aerial parts, valerian (Valeriana officinalis) leaves and little-leaf linden (Tilia cordata) flowers. PC-1 extract exhibited the strongest anti-PLO and antihemolytic effects in vitro compared to those of the individual plants and other compositions tested. Using luciferase reporter assay and Western blotting in HepG2 human hepatocellular carcinoma cells, we found that PC-1 extract activated the Nrf2/antioxidant response element signaling pathway more effectively than the extracts of other phytocompositions. Importantly, oral administration of PC-1 extract (100-200 mg/kg) markedly ameliorated liver injury in rats acutely or chronically intoxicated by carbon tetrachloride. This was evidenced by improved liver histology, blood chemistry parameters, and microsomal LPO status and superoxide dismutase activity. In addition, treatment with PC-1 extract salvaged the osmotic resistance of erythrocytes in carbon tetrachloride-intoxicated rats. Collectively, these data support the strategy of in-vitro plant selection for developing efficacious tissue-protective phytocompositions.
Collapse
Affiliation(s)
- Araylim N Aralbaeva
- Department of Biotechnology, Faculty of Food Production, Almaty Technological University, Almaty, 050012, Kazakhstan
| | - Aigul T Mamataeva
- Department of Biotechnology, Faculty of Food Production, Almaty Technological University, Almaty, 050012, Kazakhstan
| | - Nazgul I Zhaparkulova
- Department of Biophysics and Biomedicine, Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty, 480078, Kazakhstan
| | - Raisa S Utegalieva
- Department of Biotechnology, Faculty of Food Production, Almaty Technological University, Almaty, 050012, Kazakhstan
| | - Marina Khanin
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer Sheva, 8410501, Israel
| | - Michael Danilenko
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer Sheva, 8410501, Israel.
| | - Maira K Murzakhmetova
- Department of Biophysics and Biomedicine, Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty, 480078, Kazakhstan
| |
Collapse
|
43
|
Imipramine blue sensitively and selectively targets FLT3-ITD positive acute myeloid leukemia cells. Sci Rep 2017; 7:4447. [PMID: 28667329 PMCID: PMC5493614 DOI: 10.1038/s41598-017-04796-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 04/20/2017] [Indexed: 01/04/2023] Open
Abstract
Aberrant cytokine signaling initiated from mutant receptor tyrosine kinases (RTKs) provides critical growth and survival signals in high risk acute myeloid leukemia (AML). Inhibitors to FLT3 have already been tested in clinical trials, however, drug resistance limits clinical efficacy. Mutant receptor tyrosine kinases are mislocalized in the endoplasmic reticulum (ER) of AML and play an important role in the non-canonical activation of signal transducer and activator of transcription 5 (STAT5). Here, we have tested a potent new drug called imipramine blue (IB), which is a chimeric molecule with a dual mechanism of action. At 200–300 nM concentrations, IB is a potent inhibitor of STAT5 through liberation of endogenous phosphatase activity following NADPH oxidase (NOX) inhibition. However, at 75–150 nM concentrations, IB was highly effective at killing mutant FLT3-driven AML cells through a similar mechanism as thapsigargin (TG), involving increased cytosolic calcium. IB also potently inhibited survival of primary human FLT3/ITD+ AML cells compared to FLT3/ITDneg cells and spared normal umbilical cord blood cells. Therefore, IB functions through a mechanism involving vulnerability to dysregulated calcium metabolism and the combination of fusing a lipophilic amine to a NOX inhibiting dye shows promise for further pre-clinical development for targeting high risk AML.
Collapse
|
44
|
Hassan HE, Keita JA, Narayan L, Brady SM, Frederick R, Carlson S, C Glass K, Natesan S, Buttolph T, Fandy TE. The combination of dimethoxycurcumin with DNA methylation inhibitor enhances gene re-expression of promoter-methylated genes and antagonizes their cytotoxic effect. Epigenetics 2016; 11:740-749. [PMID: 27588609 DOI: 10.1080/15592294.2016.1226452] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Curcumin and its analogs exhibited antileukemic activity either as single agent or in combination therapy. Dimethoxycurcumin (DMC) is a more metabolically stable curcumin analog that was shown to induce the expression of promoter-methylated genes without reversing DNA methylation. Accordingly, co-treatment with DMC and DNA methyltransferase (DNMT) inhibitors could hypothetically enhance the re-expression of promoter-methylated tumor suppressor genes. In this study, we investigated the cytotoxic effects and epigenetic changes associated with the combination of DMC and the DNMT inhibitor decitabine (DAC) in primary leukemia samples and cell lines. The combination demonstrated antagonistic cytotoxic effects and was minimally cytotoxic to primary leukemia cells. The combination did not affect the metabolic stability of DMC. Although the combination enhanced the downregulation of nuclear DNMT proteins, the hypomethylating activity of the combination was not increased significantly compared to DAC alone. On the other hand, the combination significantly increased H3K27 acetylation (H3K27Ac) compared to the single agents near the promoter region of promoter-methylated genes. Furthermore, sequential chromatin immunoprecipitation (ChIP) and DNA pyrosequencing of the chromatin-enriched H3K27Ac did not show any significant decrease in DNA methylation compared to other regions. Consequently, the enhanced induction of promoter-methylated genes by the combination compared to DAC alone is mediated by a mechanism that involves increased histone acetylation and not through potentiation of the DNA hypomethylating activity of DAC. Collectively, our results provide the mechanistic basis for further characterization of this combination in leukemia animal models and early phase clinical trials.
Collapse
Affiliation(s)
- Hazem E Hassan
- a Department of Pharmaceutical Sciences , University of Maryland , Baltimore , MD , USA.,b Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy , Helwan University , Cairo , Egypt
| | - Jean-Arnaud Keita
- c Department of Pharmaceutical Sciences , Albany College of Pharmacy (Vermont Campus) , Colchester , VT , USA
| | - Lawrence Narayan
- c Department of Pharmaceutical Sciences , Albany College of Pharmacy (Vermont Campus) , Colchester , VT , USA
| | - Sean M Brady
- c Department of Pharmaceutical Sciences , Albany College of Pharmacy (Vermont Campus) , Colchester , VT , USA
| | - Richard Frederick
- c Department of Pharmaceutical Sciences , Albany College of Pharmacy (Vermont Campus) , Colchester , VT , USA
| | - Samuel Carlson
- c Department of Pharmaceutical Sciences , Albany College of Pharmacy (Vermont Campus) , Colchester , VT , USA
| | - Karen C Glass
- c Department of Pharmaceutical Sciences , Albany College of Pharmacy (Vermont Campus) , Colchester , VT , USA
| | - Senthil Natesan
- d Department of Experimental and Systems Pharmacology , Washington State University , Spokane , WA , USA
| | - Thomm Buttolph
- e Department of Neurological Sciences , University of Vermont , Burlington , VT , USA
| | - Tamer E Fandy
- c Department of Pharmaceutical Sciences , Albany College of Pharmacy (Vermont Campus) , Colchester , VT , USA
| |
Collapse
|
45
|
Bahri S, Jameleddine S, Shlyonsky V. Relevance of carnosic acid to the treatment of several health disorders: Molecular targets and mechanisms. Biomed Pharmacother 2016; 84:569-582. [PMID: 27694001 DOI: 10.1016/j.biopha.2016.09.067] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 08/29/2016] [Accepted: 09/18/2016] [Indexed: 12/14/2022] Open
Abstract
Carnosic acid is a phenolic diterperne compound found in abundance in sage and rosemary, which are both widely used in traditional medicine. Research over the past decade indicates that carnosic acid has multiple bioactive properties including antioxidant, anti-inflammatory and anticancer activities among others. This review summarizes the current in vitro and in vivo data about the efficacy of carnosic acid in the prevention or treatment of various experimental health disorders. The analysis of the literature allows an insight into the participation of numerous signaling pathways modulated by carnosic acid, into its synergistic potential and, thus, into the divergence in cellular mechanisms of action of this molecule.
Collapse
Affiliation(s)
- Sana Bahri
- Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, La Rabta 1007, Tunis, Tunisia; Laboratory of Physiopathology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.
| | - Saloua Jameleddine
- Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, La Rabta 1007, Tunis, Tunisia
| | - Vadim Shlyonsky
- Laboratory of Physiopathology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
46
|
The Natural Occurring Compounds Targeting Endoplasmic Reticulum Stress. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:7831282. [PMID: 27563337 PMCID: PMC4987485 DOI: 10.1155/2016/7831282] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 07/04/2016] [Indexed: 12/14/2022]
Abstract
ER stress has been implicated in pathophysiological development of many diseases. Persistent overwhelming stimuli trigger ER stress to initiate apoptosis, autophagy, and cell death. IRE1-JNK and eIF2α-CHOP signaling pathways are the two important players of ER stress, which is also modulated by ROS production, calcium disturbance, and inflammatory factors. ER stress has been developed as a novel strategy for diseases management. Recently, a vast of research focuses on the natural occurring compounds targeting ER stress, which results in medical benefits to human diseases. These small reported molecules mainly include polyphenols, alkaloids, and saponins. Many of them have been developed for use in clinical applications. To better understand the pharmacological mechanism of these molecules in ER stress in diseases, efforts have been made to discover and deliver medical merits. In this paper, we will summarize the natural occurring compounds targeting ER stress.
Collapse
|
47
|
Zhamanbayeva GT, Aralbayeva AN, Murzakhmetova MK, Tuleukhanov ST, Danilenko M. Cooperative antiproliferative and differentiation-enhancing activity of medicinal plant extracts in acute myeloid leukemia cells. Biomed Pharmacother 2016; 82:80-9. [PMID: 27470342 DOI: 10.1016/j.biopha.2016.04.062] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 04/28/2016] [Accepted: 04/28/2016] [Indexed: 12/18/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematopoietic malignancy with poor prognosis and limited treatment options. Sea buckthorn (Hippophae rhamnoides) berries, dog rose (Rosa canina) rosehips, and garden sage (Salvia officinalis) and oregano (Origanum vulgare) aerial parts are widely used in traditional medicine and exhibit antitumor effects in preclinical models. However, these plants remain scarcely tested for antileukemic activity. Here, we show that their water-ethanol leaf extracts reduced the growth and viability of AML cells and, at non-cytotoxic doses, potentiated cell differentiation induced by a low concentration of 1α,25-dihydroxyvitamin D3, the hormonal form of vitamin D, in a cell type-dependent manner. The latter effect was accompanied by upregulation of the vitamin D receptor protein components and its transcriptional activity. Furthermore, at minimally effective doses the extracts cooperated with one another to produce marked cytostatic effects associated with a partial S-phase arrest and a modest induction of apoptosis. In contrast, these combinations only slightly affected the growth and viability of proliferating normal human peripheral blood mononuclear cells. In addition, the extracts strongly inhibited microsomal lipid peroxidation and protected normal erythrocytes against hypoosmotic shock. Our results suggest that further exploration of the enhanced antileukemic effects of the combinations tested here may lead to the development of alternative therapeutic and preventive approaches against AML.
Collapse
Affiliation(s)
- Gulzhan T Zhamanbayeva
- Department of Biophysics and Biomedicine, Al-Farabi Kazakh National University, Almaty 480078, Kazakhstan
| | - Araylim N Aralbayeva
- Laboratory of Membrane Physiology, Institute of Human and Animal Physiology, Almaty 050060, Kazakhstan
| | - Maira K Murzakhmetova
- Laboratory of Membrane Physiology, Institute of Human and Animal Physiology, Almaty 050060, Kazakhstan
| | - Sultan T Tuleukhanov
- Department of Biophysics and Biomedicine, Al-Farabi Kazakh National University, Almaty 480078, Kazakhstan
| | - Michael Danilenko
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel.
| |
Collapse
|