1
|
Sivri NS, Tetikoğlu S, Kolayli S, Farooqi AA, Çelik Uzuner S. Anti-metastatic Effects of Bee Venom and Melittin in Breast Cancer Cells by Upregulation of BRMS1 and DRG1 Genes. Chem Biol Drug Des 2024; 104:e14637. [PMID: 39396919 DOI: 10.1111/cbdd.14637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024]
Abstract
Apitherapy has started to gain tremendous recognition because of extraordinary pharmacological importance of honeybee-related ingredients and their derivatives. There has been a renewed interest in the bee venom-based therapies. Interdisciplinary researchers are studying the chemistry and translational value of venom for effective cancer treatment. Bee venom and its major component, melittin, are cytotoxic in cancer cells. In this study, MTT and scratch assays were performed for analysis of melittin-mediated antimetastatic effects. QPCR was used for expression profiling of metastasis-related genes. Three anti-metastatic genes (BRMS1, DRG1, and KAI1/CD82) were studied for the first time after bee venom and melittin treatment in MDA-MB-231 breast cancer cells compared with normal breast cells, and two prometastatic genes (EGFR and WNT7B) were also examined. KAI1/CD82 and BRMS1 are the negative regulators of EGFR. WNT7B is a negative regulator of KAI1/CD82. Selective cytotoxicity of bee venom and melittin was found to be higher as compared to cisplatin. Melittin induced an increase in the expression of BRMS1 and DRG1, whereas bee venom upregulated DRG1 and KAI1/CD82 expression in breast cancer. WNT7B was downregulated in bee venom-treated breast cancer cells. Results suggested that bee venom/melittin exerted antimetastatic effects primarily through upregulation of BRMS1, DRG1, and KAI1/CD82, and downregulation of WNT7B.
Collapse
Affiliation(s)
- Nur Sena Sivri
- Department of Molecular Biology and Genetics, Karadeniz Technical University, Trabzon, Turkey
| | - Sinan Tetikoğlu
- Department of Molecular Biology and Genetics, Karadeniz Technical University, Trabzon, Turkey
| | - Sevgi Kolayli
- Department of Biochemistry, Faculty of Science, Karadeniz Technical University, Trabzon, Turkey
| | | | - Selcen Çelik Uzuner
- Department of Molecular Biology and Genetics, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
2
|
Kim BY, Lee KS, Jin BR. Antioxidant Activity and Mechanism of Action of Amwaprin: A Protein in Honeybee ( Apis mellifera) Venom. Antioxidants (Basel) 2024; 13:469. [PMID: 38671917 PMCID: PMC11047345 DOI: 10.3390/antiox13040469] [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: 03/19/2024] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Bee venom contains several bioactive components, including enzymatic and non-enzymatic proteins. There is increasing interest in the bioactive components of bee venom since they have exhibited various pharmacological effects. Recently, Apis mellifera waprin (Amwaprin) was identified as a novel protein in Apis mellifera (honeybee) venom and characterized as an antimicrobial agent. Herein, the novel biological function of Amwaprin as an antioxidant is described. In addition, the antioxidant effects of Amwaprin in mammalian cells were investigated. Amwaprin inhibited the growth of, oxidative stress-induced cytotoxicity, and inflammatory response in mammalian NIH-3T3 cells. Amwaprin decreased caspase-3 activity during oxidative stress and exhibited protective activity against oxidative stress-induced cell apoptosis in NIH-3T3 and insect Sf9 cells. The mechanism underlying the cell protective effect of Amwaprin against oxidative stress is due to its direct binding to the cell membrane. Furthermore, Amwaprin demonstrated radical-scavenging activity and protected against oxidative DNA damage. These results suggest that the antioxidant capacity of Amwaprin is attributed to the synergistic effects of its radical-scavenging action and cell shielding, indicating its novel role as an antioxidant agent.
Collapse
Affiliation(s)
| | - Kwang-Sik Lee
- College of Natural Resources and Life Science, Dong-A University, Busan 49315, Republic of Korea;
| | - Byung-Rae Jin
- College of Natural Resources and Life Science, Dong-A University, Busan 49315, Republic of Korea;
| |
Collapse
|
3
|
Dinu M, Tatu AL, Cocoș DI, Nwabudike LC, Chirilov AM, Stefan CS, Earar K, Dumitriu Buzia O. Natural Sources of Therapeutic Agents Used in Skin Conditions. Life (Basel) 2024; 14:492. [PMID: 38672762 PMCID: PMC11051086 DOI: 10.3390/life14040492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Skin conditions are numerous and often have a major impact on patients' quality of life, and effective and safe treatment is very important. The conventional drugs used for skin diseases are usually corticosteroids and antimicrobial products that can induce various side effects, especially with long-term use, which is why researchers are studying alternatives, especially biologically active natural products. Three products caught our attention: bee venom (BV), due to reported experimental results showing anti-inflammatory, antibacterial, antiviral, antioxidant, antimycotic, and anticancer effects, Ficus carica (FC) due to its demonstrated antioxidant, antibacterial, and anti-inflammatory action, and finally Geranium essential oil (GEO), with proven antifungal, antibacterial, anti-inflammatory, and antioxidant effects. Following a review of the literature, we produced this paper, which presents a review of the potential therapeutic applications of the three products in combating various skin conditions and for skin care, because BV, FC, and GEO have common pharmacological actions (anti-inflammatory, antibacterial, and antioxidant). We also focused on studying the safety of the topical use of BV, FC, and GEO, and new approaches to this. This paper presents the use of these natural therapeutic agents to treat patients with conditions such as vitiligo, melasma, and melanoma, as well as their use in treating dermatological conditions in patients with diabetes.
Collapse
Affiliation(s)
- Monica Dinu
- Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (M.D.); (C.S.S.); (K.E.); (O.D.B.)
| | - Alin Laurențiu Tatu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania;
- Dermatology Department, “Sf. Cuvioasa Parascheva” Clinical Hospital of Infectious Diseases, 800179 Galati, Romania
- Multidisciplinary Integrative Center for Dermatologic Interface Research MIC-DIR, 800010 Galati, Romania
| | - Dorin Ioan Cocoș
- Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (M.D.); (C.S.S.); (K.E.); (O.D.B.)
| | | | - Ana Maria Chirilov
- Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (M.D.); (C.S.S.); (K.E.); (O.D.B.)
| | - Claudia Simona Stefan
- Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (M.D.); (C.S.S.); (K.E.); (O.D.B.)
| | - Kamel Earar
- Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (M.D.); (C.S.S.); (K.E.); (O.D.B.)
| | - Olimpia Dumitriu Buzia
- Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (M.D.); (C.S.S.); (K.E.); (O.D.B.)
| |
Collapse
|
4
|
Jafari Z, Sadeghi S, Dehaghi MM, Bigham A, Honarmand S, Tavasoli A, Hoseini MHM, Varma RS. Immunomodulatory activities and biomedical applications of melittin and its recent advances. Arch Pharm (Weinheim) 2024; 357:e2300569. [PMID: 38251938 DOI: 10.1002/ardp.202300569] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024]
Abstract
Melittin (MLT), a peptide containing 26 amino acids, is a key constituent of bee venom. It comprises ∼40%-60% of the venom's dry weight and is the main pricing index for bee venom, being the causative factor of pain. The unique properties of MLT extracted from bee venom have made it a very valuable active ingredient in the pharmaceutical industry as this cationic and amphipathic peptide has propitious effects on human health in diverse biological processes. It has the ability to strongly impact the membranes of cells and display hemolytic activity with anticancer characteristics. However, the clinical application of MLT has been limited by its severe hemolytic activity, which poses a challenge for therapeutic use. By employing more efficient mechanisms, such as modifying the MLT sequence, genetic engineering, and nano-delivery systems, it is anticipated that the limitations posed by MLT can be overcome, thereby enabling its wider application in therapeutic contexts. This review has outlined recent advancements in MLT's nano-delivery systems and genetically engineered cells expressing MLT and provided an overview of where the MLTMLT's platforms are and where they will go in the future with the challenges ahead. The focus is on exploring how these approaches can overcome the limitations associated with MLT's hemolytic activity and improve its selectivity and efficacy in targeting cancer cells. These advancements hold promise for the creation of innovative and enhanced therapeutic approaches based on MLT for the treatment of cancer.
Collapse
Affiliation(s)
- Zohreh Jafari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Sadeghi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Mirzarazi Dehaghi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ashkan Bigham
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy (IPCB-CNR), Naples, Italy
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
| | - Shokouh Honarmand
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Afsaneh Tavasoli
- Department of Biotechnology, Faculty of Pharmacy, Alborz University of Medical Sciences, Karaj, Iran
| | - Mostafa Haji Molla Hoseini
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rajender S Varma
- Department of Chemistry, Centre of Excellence for Research in Sustainable Chemistry, Federal University of São Carlos, São Carlos, Brazil
| |
Collapse
|
5
|
Todorova T, Boyadzhiev K, Dimitrov M, Parvanova P. Bee venom genotoxicity on Saccharomyces cerevisiae cells - The role of mitochondria and YAP1 transcription factor. Toxicology 2024; 503:153768. [PMID: 38442839 DOI: 10.1016/j.tox.2024.153768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/22/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
The present work aims to clarify the genotype differences of a model organism Saccharomyces cerevisiae in response to bee venom. The study evaluated various endpoints including cell survival, induction of physiologically active superoxide anions, mitotic gene conversion, mitotic crossing-over, reverse mutations, DNA double-strand breaks, and Ty1 retrotransposition. The role of the intact mitochondria and the YAP1 transcription factor was also evaluated. Our results indicate a genotype-specific response. The first experimental evidence has been provided that bee venom induces physiologically active superoxide anions and DNA double-strand breaks in S. cerevisiae. The lack of oxidative phosphorylation due to disrupted or missing mitochondrial DNA reduces but not diminishes the cytotoxicity of bee venom. The possible modes of action could be considered direct damage to membranes (cytotoxic effect) and indirect damage to DNA through oxidative stress (genotoxic effect). YAP1 transcription factor was not found to be directly involved in cell defense against bee venom treatment.
Collapse
Affiliation(s)
- Teodora Todorova
- Institute of Biodiversity and Ecosystem Research, Bulgarian Academy of Sciences, 2 Gagarin str., Sofia 1113, Bulgaria.
| | - Krassimir Boyadzhiev
- Institute of Biodiversity and Ecosystem Research, Bulgarian Academy of Sciences, 2 Gagarin str., Sofia 1113, Bulgaria
| | - Martin Dimitrov
- Institute of Biodiversity and Ecosystem Research, Bulgarian Academy of Sciences, 2 Gagarin str., Sofia 1113, Bulgaria
| | - Petya Parvanova
- Institute of Biodiversity and Ecosystem Research, Bulgarian Academy of Sciences, 2 Gagarin str., Sofia 1113, Bulgaria
| |
Collapse
|
6
|
Kim JH, Kim TY, Goo B, Park Y. Bee Venom Stimulates Growth Factor Release from Adipose-Derived Stem Cells to Promote Hair Growth. Toxins (Basel) 2024; 16:84. [PMID: 38393162 PMCID: PMC10892121 DOI: 10.3390/toxins16020084] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Limited evidence suggests that stimulating adipose-derived stem cells (ASCs) indirectly promotes hair growth. We examined whether bee venom (BV) activated ASCs and whether BV-induced hair growth was facilitated by enhanced growth factor release by ASCs. The induction of the telogen-to-anagen phase was studied in mice. The underlying mechanism was investigated using organ cultures of mouse vibrissa hair follicles. When BV-treated ASCs were injected subcutaneously into mice, the telogen-to-anagen transition was accelerated and, by day 14, the hair weight increased. Quantitative polymerase chain reaction (qPCR) revealed that BV influenced the expression of several molecules, including growth factors, chemokines, channels, transcription factors, and enzymes. Western blot analysis was employed to verify the protein expression levels of extracellular-signal-regulated kinase (ERK) and phospho-ERK. Both the Boyden chamber experiment and scratch assay confirmed the upregulation of cell migration by BV. Additionally, ASCs secreted higher levels of growth factors after exposure to BV. Following BV therapy, the gene expression levels of alkaline phosphatase (ALP), fibroblast growth factor (FGF)-1 and 6, endothelial cell growth factor, and platelet-derived growth factor (PDGF)-C were upregulated. The findings of this study suggest that bee venom can potentially be utilized as an ASC-preconditioning agent for hair regeneration.
Collapse
Affiliation(s)
- Jung Hyun Kim
- Department of Acupuncture & Moxibustion, Kyung Hee University Hospital at Gangdong, 892, Dongnam-ro, Gangdong-gu, Seoul 05278, Republic of Korea
| | - Tae Yoon Kim
- Department of Traditional Korean Medicine Practice, Jaseng Medical Foundation, 538, Gangnam-daero, Gangnam-gu, Seoul 06110, Republic of Korea
| | - Bonhyuk Goo
- Department of Acupuncture & Moxibustion, Kyung Hee University Hospital at Gangdong, 892, Dongnam-ro, Gangdong-gu, Seoul 05278, Republic of Korea
| | - Yeoncheol Park
- Department of Acupuncture & Moxibustion Medicine, Kyung Hee University College of Korean Medicine, Kyung Hee University Hospital at Gangdong, 26, Kyungheedae-ro 4-gil, Dongdaemun-gu, Seoul 02453, Republic of Korea
| |
Collapse
|
7
|
Obeidat M, Al-Khraisat IF, Jaradat DMM, Ghanim BY, Abdallah QM, Arqoub DA, Sabbah D, Al-Sanabra OM, Arafat T, Qinna NA. Mellitin peptide quantification in seasonally collected crude bee venom and its anticancer effects on myelogenous K562 human leukaemia cell line. BMC Complement Med Ther 2023; 23:132. [PMID: 37098530 PMCID: PMC10127481 DOI: 10.1186/s12906-023-03897-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/24/2023] [Indexed: 04/27/2023] Open
Abstract
BACKGROUND Apitherapy is an emerging field in cancer research, particularly in developing communities. The potency of Melittin (MEL), a major constituent in bee venom is accounted for the cytotoxic capacity against cancer cells. It is postulated that the genotype of bees and the time of venom collection influences its specific activity against certain types of cancer. METHOD Hereby, Jordanian crude bee venom (JCBV) was collected during different seasons of the year, specifically spring, summer and autumn and investigated for in vitro antitumour effects. Venom collected during springtime comprised the highest quantity of MEL in comparison to venom collected some other time. Springtime-collected JCBV extract and MEL were tested on an immortal myelogenous leukaemia cell line, namely K562 leukemic cells. Treated cells were examined for cell modality via flow cytometry analysis and cell death mediating gene expressions. RESULTS Springtime-collected JCBV extract and MEL showed an IC50 of 3.7 ± 0.37 μg/ml and 1.84 ± 0.75 μg/ml, respectively. In comparison to JCBV and positive control, MEL-treated cells exhibited late apoptotic death with a moderate cellular arrest at G0/G1 and an increase of cell number at G2/M phase. Expression of NF-κB/MAPK14 axis was inhibited in MEL and JCBV-treated cells, as well as expression of c-MYC and CDK4. Moreover, marked upregulation in ABL1, JUN and TNF was observed. In conclusion, springtime-collected JCBV showed the highest content of MEL while both JCBV and pure MEL showed apoptotic, necrotic, and cell cycle arrest efficiency against K562 leukemic cells. CONCLUSION Integration of bee venom in chemotherapy needs more investigation and should be carefully translated into clinical use. During such translation, the correlation of bee genotype, collection time and concentration of MEL in CBV should be profiled.
Collapse
Affiliation(s)
- Maher Obeidat
- Department of Medical Laboratory Analysis, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Ihab F Al-Khraisat
- Department of Medical Laboratory Analysis, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Da'san M M Jaradat
- Department of Chemistry, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Bayan Y Ghanim
- Department of Pharmacology and Biomedical Sciences, University of Petra Pharmaceutical Center (UPPC), Faculty of Pharmacy and Medical Sciences, University of Petra, P.O. Box 961343, Amman, Jordan
| | - Qasem M Abdallah
- Department of Pharmacology and Biomedical Sciences, University of Petra Pharmaceutical Center (UPPC), Faculty of Pharmacy and Medical Sciences, University of Petra, P.O. Box 961343, Amman, Jordan
| | - Duaa Abu Arqoub
- Department of Pharmacology and Biomedical Sciences, University of Petra Pharmaceutical Center (UPPC), Faculty of Pharmacy and Medical Sciences, University of Petra, P.O. Box 961343, Amman, Jordan
| | - Duaa Sabbah
- Department of Pharmacology and Biomedical Sciences, University of Petra Pharmaceutical Center (UPPC), Faculty of Pharmacy and Medical Sciences, University of Petra, P.O. Box 961343, Amman, Jordan
| | - Ola M Al-Sanabra
- Department of Medical Laboratory Analysis, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Tawfiq Arafat
- Jordan Center for Pharmaceutical Research (JCPR), Amman, Jordan
| | - Nidal A Qinna
- Department of Pharmacology and Biomedical Sciences, University of Petra Pharmaceutical Center (UPPC), Faculty of Pharmacy and Medical Sciences, University of Petra, P.O. Box 961343, Amman, Jordan.
| |
Collapse
|
8
|
Ertilav K, Nazıroğlu M. Honey bee venom melittin increases the oxidant activity of cisplatin and kills human glioblastoma cells by stimulating the TRPM2 channel. Toxicon 2023; 222:106993. [PMID: 36528210 DOI: 10.1016/j.toxicon.2022.106993] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/30/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022]
Abstract
Melittin (MLT) treatment is believed to enhance tumor cell death, apoptotic, and oxidative cytotoxic effects of cisplatin (CSP) via the modulation of Ca2+ channels in several cancer lines. The activation of TRPM2 mediated anticancer and CSP resistance actions via mitochondrial Ca2+ and Zn2+ accumulation-induced mitochondrial reactive free oxygen species (MitSOX) in the glioblastoma cells. The aim was to elucidate the effects of CSP and MLT combination via the TRPM2 stimulation on the tumor cell viability, cell number, cell death (propidium iodide/Hoechst rate), apoptosis, and MitSOX levels in the DBTRG-05MG cells. In the DBTRG-05MG cells, we induced four groups as control, MLT (2.5 μg/ml for 24 h), CSP (25 μM for 24 h), and CSP + MLT. The CSP-induced intracellular Ca2+ influxes to the TRPM2 activation were increased in the cells from coming H2O2 and ADP-Ribose. The influxes were decreased in the cells by the incubations of TRPM2 antagonists (ACA and carvacrol). The incubation of CSP increased the parameters of intracellular Ca2+ responses, mitochondria function, cytosolic free Zn2+ accumulation, apoptosis (caspase -3, -8, and -9), and MitSOX generation in the tumor cells. After the treatment of MLT with/without CSP, the parameters were further increased in the cells. In conclusion, the treatment of MLT increased the anticancer, tumor cell death, apoptotic, and oxidant effects of CSP in the glioblastoma tumor cells via activating the TRPM2. As a result, TRPM2 stimulation by MLT may be utilized as a successful agent in the CSP treatment of glioblastoma tumors.
Collapse
Affiliation(s)
- Kemal Ertilav
- Department of Neurosurgery, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Mustafa Nazıroğlu
- Neuroscience Research Center, Suleyman Demirel University, Isparta, Turkey; BSN Health, Analysis and Innovation Ltd., Goller Bolgesi Teknokenti, Isparta, Turkey.
| |
Collapse
|
9
|
Varol A, Sezen S, Evcimen D, Zarepour A, Ulus G, Zarrabi A, Badr G, Daştan SD, Orbayoğlu AG, Selamoğlu Z, Varol M. Cellular targets and molecular activity mechanisms of bee venom in cancer: recent trends and developments. TOXIN REV 2022; 41:1382-1395. [DOI: 10.1080/15569543.2021.2024576] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 12/27/2021] [Accepted: 12/27/2021] [Indexed: 12/24/2022]
Affiliation(s)
- Ayşegül Varol
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Serap Sezen
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
- Faculty of Engineering and Natural Science, Sabanci University, Istanbul, Turkey
| | - Dilhan Evcimen
- Department of Molecular Biology and Genetics, Faculty of Science, Kotekli Campus, Mugla Sitki Kocman University, Mugla, Turkey
| | - Atefeh Zarepour
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, Turkey
| | - Gönül Ulus
- Department of Biology, Faculty of Science, Ege University, Izmir, Turkey
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, Turkey
| | - Gamal Badr
- Department of Zoology, Faculty of Science, Laboratory of Immunology, Assiut University, Assiut, Egypt
| | - Sevgi Durna Daştan
- Department of Biology, Faculty of Science, Sivas Cumhuriyet University, Sivas, Turkey
| | - Asya Gülistan Orbayoğlu
- Department of Molecular Biology and Genetics, Faculty of Science, Kotekli Campus, Mugla Sitki Kocman University, Mugla, Turkey
| | - Zeliha Selamoğlu
- Department Medical Biology, Faculty of Medicine, Nigde Ömer Halisdemir University, Nigde, Turkey
| | - Mehmet Varol
- Department of Molecular Biology and Genetics, Faculty of Science, Kotekli Campus, Mugla Sitki Kocman University, Mugla, Turkey
| |
Collapse
|
10
|
K Bakhiet E, A M Hussien H, Elshehaby M. Apis mellifera Venom Inhibits Bacterial and Fungal Pathogens in vitro. Pak J Biol Sci 2022; 25:875-884. [PMID: 36404740 DOI: 10.3923/pjbs.2022.875.884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
<b>Background and Objective:</b> Bacterial and fungal infections are major public health problems. Emerging of drug-resistant microbial strains urges the need for the development of alternative untraditional antimicrobial agents. Bee venom is a rich source of secondary metabolites and antimicrobial agents. In this study, the antimicrobial and antifungal potential of <i>Apis mellifera</i> BV (<i>Am</i>BV) against some medically important bacterial and fungal pathogens was investigated. <b>Materials and Methods:</b> Broth microdilution method and Colony Forming Unit (CFU) assay were used to screen the antibacterial potential of <i>Am</i>BV. Similarly, the antifungal activity of <i>Am</i>BV was evaluated using the agar-well diffusion assay. Moreover, the minimum inhibitory concentration (MIC) values of <i>Am</i>BV against tested microorganisms were determined. <b>Results:</b> <i>Am</i>BV significantly inhibited bacterial and fungal growth. The MIC values of <i>Am</i>BV were 15.625, 31.25, 7.8, 7.8 μg mL<sup></sup><sup>1</sup> against <i>Escherichia coli</i> ATCC 8739, <i>Staphylococcus aureus</i> ATCC 6538P, <i>Serratia marcescens</i> AUH 98 and <i>Streptococcus mutans</i> ATCC 25175, respectively. Similarly, <i>Am</i>BV at concentrations of 300 and 600 μg mL<sup></sup><sup>1</sup> significantly inhibited the growth of <i>Aspergillus niger</i> ATCC 16404, <i>Alternaria alternata</i> MLBM09, <i>Fusarium oxysporum </i>MLBM212 and <i>Aspergillus flavus. </i><b>Conclusion:</b> These results indicated that<i> Am</i>BV could be used in future preclinical and clinical studies to develop cost-effective and efficient antibacterial and antifungal agents. Moreover, this study presents <i>Am</i>BV as an efficient alternative antimicrobial agent against medically important pathogens.
Collapse
|
11
|
MnO2-melittin nanoparticles serve as an effective anti-tumor immunotherapy by enhancing systemic immune response. Biomaterials 2022; 288:121706. [PMID: 35953328 DOI: 10.1016/j.biomaterials.2022.121706] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/02/2022] [Accepted: 07/29/2022] [Indexed: 01/08/2023]
|
12
|
Schistosomicidal efficacy of bee venom-loaded pluronic F127 nanomicelles in S. mansoni infected CD1 mice. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
13
|
Chemical, Cytotoxic, and Anti-Inflammatory Assessment of Honey Bee Venom from Apis mellifera intermissa. Antibiotics (Basel) 2021; 10:antibiotics10121514. [PMID: 34943726 PMCID: PMC8698958 DOI: 10.3390/antibiotics10121514] [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: 11/05/2021] [Revised: 11/23/2021] [Accepted: 12/08/2021] [Indexed: 12/03/2022] Open
Abstract
The venom from Apis mellifera intermissa, the main honey bee prevailing in Morocco, has been scarcely studied, despite its known potential for pharmacological applications. In the present work, we investigated the composition, the anti-inflammatory activity, and the venom’s cytotoxic properties from fifteen honey bee venom (HBV) samples collected in three regions: northeast, central, and southern Morocco. The chemical assessment of honey bee venom was performed using LC-DAD/ESI/MSn, NIR spectroscopy and AAS spectroscopy. The antiproliferative effect was evaluated using human tumor cell lines, including breast adenocarcinoma, non-small cell lung carcinoma, cervical carcinoma, hepatocellular carcinoma, and malignant melanoma. Likewise, we assessed the anti-inflammatory activity using the murine macrophage cell line. The study provides information on the honey bee venom subspecies’ main components, such as melittin, apamin, and phospholipase A2, with compositional variation depending on the region of collection. Contents of toxic elements such as cadmium, chromium, and plumb were detected at a concentration below 5 ppm, which can be regarded as safe for pharmaceutical use. The data presented contribute to the first study in HBV from Apis mellifera intermissa and highlight the remarkable antiproliferative and anti-inflammatory effects of HBV, suggesting it to be a candidate natural medicine to explore.
Collapse
|
14
|
Guha S, Ferrie RP, Ghimire J, Ventura CR, Wu E, Sun L, Kim SY, Wiedman GR, Hristova K, Wimley WC. Applications and evolution of melittin, the quintessential membrane active peptide. Biochem Pharmacol 2021; 193:114769. [PMID: 34543656 DOI: 10.1016/j.bcp.2021.114769] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022]
Abstract
Melittin, the main venom component of the European Honeybee, is a cationic linear peptide-amide of 26 amino acid residues with the sequence: GIGAVLKVLTTGLPALISWIKRKRQQ-NH2. Melittin binds to lipid bilayer membranes, folds into amphipathic α-helical secondary structure and disrupts the permeability barrier. Since melittin was first described, a remarkable array of activities and potential applications in biology and medicine have been described. Melittin is also a favorite model system for biophysicists to study the structure, folding and function of peptides and proteins in membranes. Melittin has also been used as a template for the evolution of new activities in membranes. Here we overview the rich history of scientific research into the many activities of melittin and outline exciting future applications.
Collapse
Affiliation(s)
- Shantanu Guha
- University of Texas Health Science Center at Houston, Department of Microbiology and Molecular Genetics, Houston, TX, USA
| | - Ryan P Ferrie
- Tulane University School of Medicine, Department of Biochemistry and Molecular Biology, New Orleans, LA, USA
| | - Jenisha Ghimire
- Tulane University School of Medicine, Department of Biochemistry and Molecular Biology, New Orleans, LA, USA
| | - Cristina R Ventura
- Seton Hall University, Department of Chemistry and Biochemistry, South Orange, NJ, USA
| | - Eric Wu
- Tulane University School of Medicine, Department of Biochemistry and Molecular Biology, New Orleans, LA, USA
| | - Leisheng Sun
- Tulane University School of Medicine, Department of Biochemistry and Molecular Biology, New Orleans, LA, USA
| | - Sarah Y Kim
- Duke University, Department of Biomedical Engineering, Durham, NC, USA
| | - Gregory R Wiedman
- Seton Hall University, Department of Chemistry and Biochemistry, South Orange, NJ, USA
| | - Kalina Hristova
- Johns Hopkins University, Department of Materials Science and Engineering, Baltimore, MD, USA.
| | - Wimley C Wimley
- University of Texas Health Science Center at Houston, Department of Microbiology and Molecular Genetics, Houston, TX, USA.
| |
Collapse
|
15
|
Bee Venom Components as Therapeutic Tools against Prostate Cancer. Toxins (Basel) 2021; 13:toxins13050337. [PMID: 34067049 PMCID: PMC8150751 DOI: 10.3390/toxins13050337] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 12/22/2022] Open
Abstract
Prostate cancer is one of the most common cancers in men. Despite the development of a variety of therapeutic agents to treat either metastatic hormone-sensitive prostate cancer, advanced prostate cancer, or nonmetastatic/metastatic castration-resistant prostate cancer, the progression or spread of the disease often cannot be avoided. Additionally, the development of resistance of prostate cancer cells to available therapeutic agents is a well-known problem. Despite extensive and cost-intensive research over decades, curative therapy for metastatic prostate cancer is still not available. Therefore, additional therapeutic agents are still needed. The animal kingdom offers a valuable source of natural substances used for the treatment of a variety of diseases. Bee venom of the honeybee is a mixture of many components. It contains proteins acting as enzymes such as phospholipase A2, smaller proteins and peptides such as melittin and apamin, phospholipids, and physiologically active amines such as histamine, dopamine, and noradrenaline. Melittin has been shown to induce apoptosis in different cancer cell lines, including prostate cancer cell lines. It also influences cell proliferation, angiogenesis, and necrosis as well as motility, migration, metastasis, and invasion of tumour cells. Hence, it represents an interesting anticancer agent. In this review article, studies about the effect of bee venom components on prostate cancer cells are discussed. An electronic literature research was performed utilising PubMed in February 2021. All scientific publications, which examine this interesting subject, are discussed. Furthermore, the different types of application of these promising substances are outlined. The studies clearly indicate that bee venom or melittin exhibited anticancer effects in various prostate cancer cell lines and in xenografts. In most of the studies, a combination of bee venom or the modified melittin with another molecule was utilised in order to avoid side effects and, additionally, to target selectively the prostate cancer cells or the surrounding tissue. The studies showed that systemic side effects and unwanted damage to healthy tissue and organs could be minimised when the anticancer drug was not activated until binding to the cancer cells or the surrounding tissue. Different targets were used, such as the matrix metalloproteinase 2, hormone receptors expressed by prostate cancer cells, the extracellular domain of PSMA, and the fibroblast activation protein occurring in the stroma of prostate cancer cells. Another approach used loaded phosphate micelles, which were cleaved by the enzyme secretory phospholipase A2 produced by prostate cancer cells. In a totally different approach, targeted nanoparticles containing the melittin gene were used for prostate cancer gene therapy. By the targeted nonviral gene delivery, the gene encoding melittin was delivered to the prostate cancer cells without systemic side effects. This review of the scientific literature reveals totally different approaches using bee venom, melittin, modified melittin, or protoxin as anticancer agents. The toxic agents acted through several different mechanisms to produce their anti-prostate cancer effects. These mechanisms are not fully understood yet and more experimental studies are necessary to reveal the complete mode of action. Nevertheless, the researchers have conducted pioneering work. Based on these results, further experimental and clinical studies about melittin and modifications of this interesting agent deriving from nature are necessary and could possibly lead to a complementary treatment option for prostate cancer.
Collapse
|
16
|
Ombredane AS, de Andrade LR, Bonadio RS, Pinheiro WO, de Azevedo RB, Joanitti GA. Melittin sensitizes skin squamous carcinoma cells to 5-fluorouracil by affecting cell proliferation and survival. Exp Dermatol 2021; 30:710-716. [PMID: 33523510 DOI: 10.1111/exd.14289] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/13/2020] [Accepted: 01/13/2021] [Indexed: 12/13/2022]
Abstract
Combined 5-fluorouracil (5-FU) and melittin (MEL) is believed to enhance cytotoxic effects on skin squamous cell carcinoma (SCC). However, the rationale underlying cytotoxicity is fundamentally important for a proper design of combination chemotherapy, and to provide translational insights for future therapeutics in the dermatology field. The aim was to elucidate the effects of 5-FU/MEL combination on the viability, proliferation and key structures of human squamous cell carcinoma (A431). Morphology, plasma membrane, DNA, mitochondria, oxidative stress, cell viability, proliferation and cell death pathways were targeted for investigation by microscopy, MTT, trypan blue assay, flow cytometry and real-time cell analysis. 5-FU/MEL (0.25 µM/0.52 µM) enhanced the cytotoxic effect in A431 cells (74.46%, p < .001) after 72 h exposure, showing greater cytotoxic effect when compared to each isolated compound (45.55% 5-FU and 61.78% MEL). The results suggest that MEL induces plasma membrane alterations that culminate in a loss of integrity at subsequent times, sensitizing the cell to 5-FU action. DNA fragmentation, S and G2/M arrest, disruption of mitochondrial metabolism, and alterations in cell morphology culminated in proliferation blockage and apoptosis. 5-FU/MEL combination design optimizes the cytotoxic effects of each drug at lower concentrations, which may represent an innovative strategy for SCC therapy.
Collapse
Affiliation(s)
- Alicia S Ombredane
- Laboratory of Bioactive Compounds and Nanobiotechnology (LBCNano, Faculty of Ceilândia, University of Brasília, Brasília, Brazil.,Post-Graduation Program in Nanoscience and Nanobiotechnology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Laise R de Andrade
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Raphael S Bonadio
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Willie O Pinheiro
- Post-Graduation Program in Sciences and Technologies in Health, Faculty of Ceilândia, University of Brasília, Brasília, Brazil
| | - Ricardo B de Azevedo
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Graziella A Joanitti
- Laboratory of Bioactive Compounds and Nanobiotechnology (LBCNano, Faculty of Ceilândia, University of Brasília, Brasília, Brazil.,Post-Graduation Program in Nanoscience and Nanobiotechnology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil.,Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil.,Post-Graduation Program in Sciences and Technologies in Health, Faculty of Ceilândia, University of Brasília, Brasília, Brazil
| |
Collapse
|
17
|
Borojeni SK, Zolfagharian H, Babaie M, Javadi I. Cytotoxic Effect of Bee ( A. mellifera) Venom on Cancer Cell Lines. J Pharmacopuncture 2020; 23:212-219. [PMID: 33408897 PMCID: PMC7772077 DOI: 10.3831/kpi.2020.23.4.212] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 01/08/2023] Open
Abstract
Objectives Nowadays cancer treatment is an important challenge in the medical world that needs better therapies. Many active secretions produced by insects such as honey bees used to discover new anticancer drugs. Bee venom (BV) has a potent anti inflammatory, anti cancer and tumor effects. The aim of present study is evaluation of anticancer effects induced by Apis mellifera venom (AmV) on cell Lines. Methods AmV was selected for study on cancer cell lines. Total protein, molecular weight and LD50 of crude venom were determined. Then, cells were grown in Dulbecco's Modified Eagle medium supplemented with 10% fetal bovine serum and 1% antibiotics. The A549, HeLa and MDA-MB-231 cell Lines were exposed by different concentration of AmV. The morphology of cells was determined and cell viability was studed by MTT assay. Evaluation of cell death was determined by and DNA fragmentation. Results The results from MTT assay showed that 3.125 µg/mL of A549, 12.5 for HeLa and 6.25 µg/mL of MDA-MB-231 killed 50% of cells (p < 0.05). Morphological analysis and the results from hoescht staining and DNA fragmentation indicated that cell death induced by AmV was significantly apoptosis. Conclusion The data showed that using lower dosage of AmV during treatment period cause inhibition of proliferation in time and dose dependant manner. Findings indicated that some ingredients of AmV have anticancer effects and with further investigation it can be used in production of anticancer drugs.
Collapse
Affiliation(s)
- Sima Khalilifard Borojeni
- Department of Venomous Animals and Antivenom Production, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Hossein Zolfagharian
- Department of Venomous Animals and Antivenom Production, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Mahdi Babaie
- Department of Venomous Animals and Antivenom Production, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran.,Young Researchers and Elites Club, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Iraj Javadi
- Department of Toxicology, Islamic Azad University, Shahreza Branch, Shahreza, Isfahan, Iran
| |
Collapse
|
18
|
Klupczynska A, Plewa S, Dereziński P, Garrett TJ, Rubio VY, Kokot ZJ, Matysiak J. Identification and quantification of honeybee venom constituents by multiplatform metabolomics. Sci Rep 2020; 10:21645. [PMID: 33303913 PMCID: PMC7729905 DOI: 10.1038/s41598-020-78740-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
Honeybee (Apis mellifera) venom (HBV) has been a subject of extensive proteomics research; however, scarce information on its metabolite composition can be found in the literature. The aim of the study was to identify and quantify the metabolites present in HBV. To gain the highest metabolite coverage, three different mass spectrometry (MS)-based methodologies were applied. In the first step, untargeted metabolomics was used, which employed high-resolution, accurate-mass Orbitrap MS. It allowed obtaining a broad overview of HBV metabolic components. Then, two targeted metabolomics approaches, which employed triple quadrupole MS, were applied to quantify metabolites in HBV samples. The untargeted metabolomics not only confirmed the presence of amines, amino acids, carbohydrates, and organic acids in HBV, but also provided information on venom components from other metabolite classes (e.g., nucleosides, alcohols, purine and pyrimidine derivatives). The combination of three MS-based metabolomics platforms facilitated the identification of 214 metabolites in HBV samples, among which 138 were quantified. The obtaining of the wide free amino acid profiles of HBV is one of the project’s achievements. Our study contributed significantly to broadening the knowledge about HBV composition and should be continued to obtain the most comprehensive metabolite profile of HBV.
Collapse
Affiliation(s)
- Agnieszka Klupczynska
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60-780, Poznan, Poland.
| | - Szymon Plewa
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60-780, Poznan, Poland
| | - Paweł Dereziński
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60-780, Poznan, Poland
| | - Timothy J Garrett
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Vanessa Y Rubio
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Zenon J Kokot
- Faculty of Health Sciences, Calisia University - Kalisz, Poland, 62-800, Kalisz, Poland
| | - Jan Matysiak
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60-780, Poznan, Poland
| |
Collapse
|
19
|
Lin TY, Hsieh CL. Clinical Applications of Bee Venom Acupoint Injection. Toxins (Basel) 2020; 12:toxins12100618. [PMID: 32992601 PMCID: PMC7601520 DOI: 10.3390/toxins12100618] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 12/29/2022] Open
Abstract
Bee venom is a complex natural mixture with various pharmaceutical properties. Among these properties, its peptides and enzymes have potential medical therapy for pain relief and inflammation. In clinical settings, this therapy has been used widely to treat diseases by injecting into acupoints. In this article, we have conducted various research from PubMed, Cochrane Library, and Clinical Key from inception of July 2020. The results revealed that bee venom therapy has been reported effective in anti-inflammatory, antiapoptosis, and analgesic effects. Moreover, bee venom acupuncture has been commonly used for clinical disorders such as Parkinson disease, neuropathic pain, Alzheimer disease, intervertebral disc disease, spinal cord injury, musculoskeletal pain, arthritis, multiple sclerosis, skin disease and cancer.
Collapse
Affiliation(s)
- Ting-Yen Lin
- Department of Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan;
| | - Ching-Liang Hsieh
- Department of Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan;
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan
- Correspondence: ; Tel.: +886-2205-3366-3128
| |
Collapse
|
20
|
Duffy C, Sorolla A, Wang E, Golden E, Woodward E, Davern K, Ho D, Johnstone E, Pfleger K, Redfern A, Iyer KS, Baer B, Blancafort P. Honeybee venom and melittin suppress growth factor receptor activation in HER2-enriched and triple-negative breast cancer. NPJ Precis Oncol 2020; 4:24. [PMID: 32923684 PMCID: PMC7463160 DOI: 10.1038/s41698-020-00129-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
Despite decades of study, the molecular mechanisms and selectivity of the biomolecular components of honeybee (Apis mellifera) venom as anticancer agents remain largely unknown. Here, we demonstrate that honeybee venom and its major component melittin potently induce cell death, particularly in the aggressive triple-negative and HER2-enriched breast cancer subtypes. Honeybee venom and melittin suppress the activation of EGFR and HER2 by interfering with the phosphorylation of these receptors in the plasma membrane of breast carcinoma cells. Mutational studies reveal that a positively charged C-terminal melittin sequence mediates plasma membrane interaction and anticancer activity. Engineering of an RGD motif further enhances targeting of melittin to malignant cells with minimal toxicity to normal cells. Lastly, administration of melittin enhances the effect of docetaxel in suppressing breast tumor growth in an allograft model. Our work unveils a molecular mechanism underpinning the anticancer selectivity of melittin, and outlines treatment strategies to target aggressive breast cancers.
Collapse
Affiliation(s)
- Ciara Duffy
- School of Human Sciences, The University of Western Australia, Perth, WA 6009 Australia.,Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Plant Energy Biology, The University of Western Australia, Perth, WA 6009 Australia.,Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia
| | - Anabel Sorolla
- Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia
| | - Edina Wang
- Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia
| | - Emily Golden
- Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia
| | - Eleanor Woodward
- Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia
| | - Kathleen Davern
- Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia.,Monoclonal Antibody (MAb) Facility, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia
| | - Diwei Ho
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009 Australia
| | - Elizabeth Johnstone
- Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia.,Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Australian Research Council Centre for Personalised Therapeutics Technologies, Perth, Australia
| | - Kevin Pfleger
- Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia.,Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Australian Research Council Centre for Personalised Therapeutics Technologies, Perth, Australia.,Dimerix Limited; Nedlands, Perth, WA 6009 Australia
| | - Andrew Redfern
- School of Medicine, The University of Western Australia, Perth, WA 6009 Australia
| | - K Swaminathan Iyer
- Monoclonal Antibody (MAb) Facility, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia
| | - Boris Baer
- Centre for Integrative Bee Research (CIBER), Department of Entomology; University of California Riverside, Riverside, CA 92521 USA
| | - Pilar Blancafort
- School of Human Sciences, The University of Western Australia, Perth, WA 6009 Australia.,Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia.,The Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| |
Collapse
|
21
|
Sorolla A, Sorolla MA, Wang E, Ceña V. Peptides, proteins and nanotechnology: a promising synergy for breast cancer targeting and treatment. Expert Opin Drug Deliv 2020; 17:1597-1613. [PMID: 32835538 DOI: 10.1080/17425247.2020.1814733] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The use of nanoparticles for breast cancer targeting and treatment has become a reality. They are safe and possess interesting peculiarities such as the unspecific accumulation into the tumor site and the possibility to activate controlled drug release as compared to free drugs. However, there are still many areas of improvement which can certainly be addressed with the use of peptide-based elements. AREAS COVERED The article reviews different preclinical strategies employing peptides and proteins in combination with nanoparticles for breast cancer targeting and treatment as well as peptide and protein-targeted encapsulated drugs, and it lists the current clinical status of therapies using peptides and proteins for breast cancer. EXPERT OPINION The conjugation of protein and peptides can improve tumor homing of nanoparticles, increase cellular penetration and attack specific drivers and vulnerabilities of the breast cancer cell to promote tumor cytotoxicity while reducing secondary effects in healthy tissues. Examples are the use of antibodies, arginylglycylaspartic acid (RGD) peptides, membrane disruptive peptides, interference peptides, and peptide vaccines. Although their implementation in the clinic has been relatively slow up to now, we anticipate great progress in the field which will translate into more efficacious and selective nanotherapies for breast cancer.
Collapse
Affiliation(s)
- Anabel Sorolla
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia , Crawley, Australia
| | - Maria Alba Sorolla
- Biomedical Research Institute (IRB Lleida), Research Group of Cancer Biomarkers , Lleida, Spain
| | - Edina Wang
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia , Crawley, Australia
| | - Valentín Ceña
- Unidad Asociada Neurodeath, Universidad De Castilla-La Mancha , Albacete, Spain.,Centro De Investigación En Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII , Madrid, Spain
| |
Collapse
|
22
|
Melittin from Apis florea Venom as a Promising Therapeutic Agent for Skin Cancer Treatment. Antibiotics (Basel) 2020; 9:antibiotics9080517. [PMID: 32823904 PMCID: PMC7460526 DOI: 10.3390/antibiotics9080517] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 02/02/2023] Open
Abstract
Melittin, a major component found in bee venom, is produced by the Apis species of the honey bee. In this study, the effect of melittin derived from Apis florea (Mel-AF), which is a wild honey bee species that is indigenous to Thailand, was investigated against human malignant melanoma (A375) cells. In this study, Mel-AF exhibited considerable potential in the anti-proliferative action of A375 cells. Subsequently, the cellular mechanism of Mel-AF that induced cell death was investigated in terms of apoptosis. As a result, gene and protein expression levels, which indicated the activation of cytochrome-c release and caspase-9 expression, eventually triggered the release of the caspase-3 executioner upon Mel-AF. We then determined that apoptosis-mediated cell death was carried out through the intrinsic mitochondrial pathway. Moreover, advanced abilities, including cell motility and invasion, were significantly suppressed. Mel-AF manipulated the actin arrangement via the trapping of stress fibers that were found underneath the membrane, which resulted in the defective actin cytoskeleton organization. Consequently, the expression of EGFR, a binding protein to F-actin, was also found to be suppressed. This outcome strongly supports the effects of Mel-AF in the inhibition of progressive malignant activity through the disruption of actin cytoskeleton-EGFR interaction and the EGFR signaling system. Thus, the findings of our current study indicate the potential usefulness of Mel-AF in cancer treatments as an apoptosis inducer and a potential actin-targeting agent.
Collapse
|
23
|
Kim A, Lee SY, Kim BY, Chung SK. Elimination of Teratogenic Human Induced Pluripotent Stem Cells by Bee Venom via Calcium-Calpain Pathway. Int J Mol Sci 2020; 21:ijms21093265. [PMID: 32380745 PMCID: PMC7246707 DOI: 10.3390/ijms21093265] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/29/2020] [Accepted: 05/02/2020] [Indexed: 12/14/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are regarded as a promising option for cell-based regenerative medicine. To obtain safe and efficient iPSC-based cell products, it is necessary to selectively eliminate the residual iPSCs prior to in vivo implantation due to the risk of teratoma formation. Bee venom (BV) has long been used in traditional Chinese medicine to treat inflammatory diseases and relieve pain, and has been shown to exhibit anti-cancer, anti-mutagenic, anti-nociceptive, and radioprotective activities. However, the potential benefits of BV in iPSC therapy, particularly its anti-teratoma activity, have not been examined. In this study, we found that BV selectively induced cell death in iPSCs, but not in iPSC-derived differentiated cells (iPSCs-Diff). BV rapidly disrupted cell membrane integrity and focal adhesions, followed by induction of apoptosis and necroptosis in iPSCs. We also found that BV remarkably enhanced intracellular calcium levels, calpain activation, and reactive oxygen speciesgeneration in iPSCs. BV treatment before in ovo grafting efficiently prevented iPSC-derived teratoma formation. In contrast, no DNA damage was observed in iPSCs-Diff following BV treatment, further demonstrating the safety of BV for use with iPSCs-Diff. Taken together, these findings show that BV has potent anti-teratoma activity by eliminating residual iPSCs, and can be used for the development of effective and safe iPSC-based cell therapies.
Collapse
Affiliation(s)
- Aeyung Kim
- Clinical Medicine Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
- Correspondence: (A.K.); (S.-K.C.); Tel.: +82-42-868-9674 (A.K.); +82-42-868-9634 (S.-K.C.)
| | - Seo-Young Lee
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea; (S.-Y.L.); (B.-Y.K.)
| | - Bu-Yeo Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea; (S.-Y.L.); (B.-Y.K.)
| | - Sun-Ku Chung
- Clinical Medicine Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
- Correspondence: (A.K.); (S.-K.C.); Tel.: +82-42-868-9674 (A.K.); +82-42-868-9634 (S.-K.C.)
| |
Collapse
|
24
|
Melittin-A Natural Peptide from Bee Venom Which Induces Apoptosis in Human Leukaemia Cells. Biomolecules 2020; 10:biom10020247. [PMID: 32041197 PMCID: PMC7072249 DOI: 10.3390/biom10020247] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/29/2020] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Bee venom is a very complex mixture produced and secreted by the honeybee (Apis mellifera). Melittin is a major component of bee venom that accounts for about 52% of its dry mass. A vast number of studies have been dedicated to the effects of melittin’s regulation of apoptosis and to the factors that induce apoptosis in various types of cancer such as breast, ovarian, prostate, lung. The latest evidence indicates its potential as a therapeutic agent in the treatment of leukaemia. The aim of our present study is to evaluate melittin’s ability to induce apoptosis in leukaemia cell lines of different origin acute lymphoblastic leukaemia (CCRF-CEM) and chronic myelogenous leukaemia (K-562). We demonstrated that melittin strongly reduced cell viability in both leukaemia cell lines but not in physiological peripheral blood mononuclear cells (PMBCs). Subsequent estimated parameters (mitochondrial membrane potential, Annexin V binding and Caspases 3/7 activity) clearly demonstrated that melittin induced apoptosis in leukaemia cells. This is a very important step for research into the development of new potential anti-leukaemia as well as anticancer therapies. Further analyses on the molecular level have been also planned (analysis of proapoptotic genes expression and DNA damages) for our next research project, which will also focus on melittin.
Collapse
|
25
|
Kurek-Górecka A, Górecki M, Rzepecka-Stojko A, Balwierz R, Stojko J. Bee Products in Dermatology and Skin Care. Molecules 2020; 25:molecules25030556. [PMID: 32012913 PMCID: PMC7036894 DOI: 10.3390/molecules25030556] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/22/2020] [Accepted: 01/26/2020] [Indexed: 12/31/2022] Open
Abstract
Honey, propolis, bee pollen, bee bread, royal jelly, beeswax and bee venom are natural products which have been used in medicine since ancient times. Nowadays, studies indicate that natural bee products can be used for skin treatment and care. Biological properties of these products are related to flavonoids they contain like: chrysin, apigenin, kaempferol, quercetin, galangin, pinocembrin or naringenin. Several pharmacological activities of phenolic acids and flavonoids, and also 10-hydroxy-trans-2-decenoic acid, which is present in royal jelly, have been reported. Royal jelly has multitude of pharmacological activities: antibiotic, antiinflammatory, antiallergenic, tonic and antiaging. Honey, propolis and pollen are used to heal burn wounds, and they possess numerous functional properties such as: antibacterial, anti-inflammatory, antioxidant, disinfectant, antifungal and antiviral. Beeswax is used for production of cosmetics and ointments in pharmacy. Due to a large number of biological activities, bee products could be considered as important ingredients in medicines and cosmetics applied to skin.
Collapse
Affiliation(s)
- Anna Kurek-Górecka
- Silesian Academy of Medical Sciences in Katowice, Mickiewicza 29, 40-085 Katowice, Poland;
- Correspondence:
| | - Michał Górecki
- Department of Drug Technology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland; (M.G.); (A.R.-S.)
| | - Anna Rzepecka-Stojko
- Department of Drug Technology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland; (M.G.); (A.R.-S.)
| | - Radosław Balwierz
- Silesian Academy of Medical Sciences in Katowice, Mickiewicza 29, 40-085 Katowice, Poland;
| | - Jerzy Stojko
- Department of Toxycology and Bioanalysis, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Ostrogórska 30, 41-200 Sosnowiec, Poland;
| |
Collapse
|
26
|
Use of Selected Carbon Nanoparticles as Melittin Carriers for MCF-7 and MDA-MB-231 Human Breast Cancer Cells. MATERIALS 2019; 13:ma13010090. [PMID: 31878020 PMCID: PMC6981792 DOI: 10.3390/ma13010090] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/13/2019] [Accepted: 12/19/2019] [Indexed: 12/16/2022]
Abstract
Despite advanced techniques in medicine, breast cancer caused the deaths of 627,000 women in 2018. Melittin, the main component of bee venom, has lytic properties for many types of cells, including cancer cells. To increase its toxic effect, carbon nanoparticles, graphene oxide, pristine graphene, and diamond were used as carriers of melittin to breast cancer cells. To date, the effects of carbon nanoparticles as carriers of melittin on cancer cells have not been studied. The present study was carried out on MCF-7 and MDA-MB-231 cell lines. The investigation consisted of structural analysis of complexes using transmission electron microscopy, zeta potential measurements, evaluation of cell morphology, assessment of cell viability and membrane integrity, investigation of reactive oxygen species production, and investigation of mitochondrial membrane potential. Cell death was examined by flow cytometry and a membrane test for 43 apoptotic proteins. The results indicate that melittin complex with nanographene oxide has a stronger toxic effect on breast cancer cells than melittin alone. Moreover, nanodiamonds can protect cells against the lytic effects of melittin. All complexes reduced, but not completely eliminated the level of necrosis, compared to melittin. Thus, results suggest that the use of carbon nanoparticles as carriers for melittin may find use in medicine in the future.
Collapse
|
27
|
An overview of the bioactive compounds, therapeutic properties and toxic effects of apitoxin. Food Chem Toxicol 2019; 134:110864. [PMID: 31574265 DOI: 10.1016/j.fct.2019.110864] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/15/2022]
|
28
|
Shaw P, Kumar N, Hammerschmid D, Privat-Maldonado A, Dewilde S, Bogaerts A. Synergistic Effects of Melittin and Plasma Treatment: A Promising Approach for Cancer Therapy. Cancers (Basel) 2019; 11:cancers11081109. [PMID: 31382579 PMCID: PMC6721819 DOI: 10.3390/cancers11081109] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 12/31/2022] Open
Abstract
Melittin (MEL), a small peptide component of bee venom, has been reported to exhibit anti-cancer effects in vitro and in vivo. However, its clinical applicability is disputed because of its non-specific cytotoxicity and haemolytic activity in high treatment doses. Plasma-treated phosphate buffered saline solution (PT-PBS), a solution rich in reactive oxygen and nitrogen species (RONS) can disrupt the cell membrane integrity and induce cancer cell death through oxidative stress-mediated pathways. Thus, PT-PBS could be used in combination with MEL to facilitate its access into cancer cells and to reduce the required therapeutic dose. The aim of our study is to determine the reduction of the effective dose of MEL required to eliminate cancer cells by its combination with PT-PBS. For this purpose, we have optimised the MEL threshold concentration and tested the combined treatment of MEL and PT-PBS on A375 melanoma and MCF7 breast cancer cells, using in vitro, in ovo and in silico approaches. We investigated the cytotoxic effect of MEL and PT-PBS alone and in combination to reveal their synergistic cytological effects. To support the in vitro and in ovo experiments, we showed by computer simulations that plasma-induced oxidation of the phospholipid bilayer leads to a decrease of the free energy barrier for translocation of MEL in comparison with the non-oxidized bilayer, which also suggests a synergistic effect of MEL with plasma induced oxidation. Overall, our findings suggest that MEL in combination with PT-PBS can be a promising combinational therapy to circumvent the non-specific toxicity of MEL, which may help for clinical applicability in the future.
Collapse
Affiliation(s)
- Priyanka Shaw
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, BE-2610 Wilrijk-Antwerp, Belgium
| | - Naresh Kumar
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, BE-2610 Wilrijk-Antwerp, Belgium.
| | - Dietmar Hammerschmid
- Laboratory of Protein Science, Proteomics & Epigenetic Signaling, Department of Biomedical Sciences, University of Antwerp, BE-2610 Wilrijk-Antwerp, Belgium
| | - Angela Privat-Maldonado
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, BE-2610 Wilrijk-Antwerp, Belgium
| | - Sylvia Dewilde
- Laboratory of Protein Science, Proteomics & Epigenetic Signaling, Department of Biomedical Sciences, University of Antwerp, BE-2610 Wilrijk-Antwerp, Belgium
| | - Annemie Bogaerts
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, BE-2610 Wilrijk-Antwerp, Belgium.
| |
Collapse
|
29
|
Lyu C, Fang F, Li B. Anti-Tumor Effects of Melittin and Its Potential Applications in Clinic. Curr Protein Pept Sci 2019; 20:240-250. [PMID: 29895240 DOI: 10.2174/1389203719666180612084615] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 04/10/2018] [Accepted: 05/21/2018] [Indexed: 02/08/2023]
Abstract
Melittin, a major component of bee venom, is a water-soluble toxic peptide of which a various biological effects have been identified to be useful in anti-tumor therapy. In addition, Melittin also has anti-parasitic, anti-bacterial, anti-viral, and anti-inflammatory activities. Therefore, it is a very attractive therapeutic candidate for human diseases. However, melittin induces extensive hemolysis, a severe side effect that dampens its future development and clinical application. Thus, studies of melittin derivatives and new drug delivery systems have been conducted to explore approaches for optimizing the efficacy of this compound, while reducing its toxicity. A number of reviews have focused on each side, respectively. In this review, we summarize the research progress on the anti-tumor effects of melittin and its derivatives, and discuss its future potential clinical applications.
Collapse
Affiliation(s)
- Can Lyu
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, Shanghai, China
| | - Fanfu Fang
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, Shanghai, China
| | - Bai Li
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, Shanghai, China
| |
Collapse
|
30
|
Kim H, Park SY, Lee G. Potential Therapeutic Applications of Bee Venom on Skin Disease and Its Mechanisms: A Literature Review. Toxins (Basel) 2019; 11:toxins11070374. [PMID: 31252651 PMCID: PMC6669657 DOI: 10.3390/toxins11070374] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/23/2019] [Accepted: 06/25/2019] [Indexed: 01/03/2023] Open
Abstract
Skin is larger than any other organ in humans. Like other organs, various bacterial, viral, and inflammatory diseases, as well as cancer, affect the skin. Skin diseases like acne, atopic dermatitis, and psoriasis often reduce the quality of life seriously. Therefore, effective treatment of skin disorders is important despite them not being life-threatening. Conventional medicines for skin diseases include corticosteroids and antimicrobial drugs, which are effective in treating many inflammatory and infectious skin diseases; however, there are growing concerns about the side effects of these therapies, especially during long-term use in relapsing or intractable diseases. Hence, many researchers are trying to develop alternative treatments, especially from natural sources, to resolve these limitations. Bee venom (BV) is an attractive candidate because many experimental and clinical reports show that BV exhibits anti-inflammatory, anti-apoptotic, anti-fibrotic, antibacterial, antiviral, antifungal, and anticancer effects. Here, we review the therapeutic applications of BV in skin diseases, including acne, alopecia, atopic dermatitis, melanoma, morphea, photoaging, psoriasis, wounds, wrinkles, and vitiligo. Moreover, we explore the therapeutic mechanisms of BV in the treatment of skin diseases and killing effects of BV on skin disease-causing pathogens, including bacteria, fungi and viruses.
Collapse
Affiliation(s)
- Haejoong Kim
- College of Korean Medicine, Dongshin University, Naju-si, Jeollanam-do 58245, Korea
| | - Soo-Yeon Park
- Department of Ophthalmology, Otolaryngology & Dermatology, College of Korean Medicine, Dongshin University, Naju-si, Jeollanam-do 58245, Korea.
| | - Gihyun Lee
- College of Korean Medicine, Dongshin University, Naju-si, Jeollanam-do 58245, Korea.
| |
Collapse
|
31
|
Lim HN, Baek SB, Jung HJ. Bee Venom and Its Peptide Component Melittin Suppress Growth and Migration of Melanoma Cells via Inhibition of PI3K/AKT/mTOR and MAPK Pathways. Molecules 2019; 24:molecules24050929. [PMID: 30866426 PMCID: PMC6429308 DOI: 10.3390/molecules24050929] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 02/28/2019] [Accepted: 03/03/2019] [Indexed: 01/28/2023] Open
Abstract
Malignant melanoma is the deadliest form of skin cancer and highly chemoresistant. Melittin, an amphiphilic peptide containing 26 amino acid residues, is the major active ingredient from bee venom (BV). Although melittin is known to have several biological activities such as anti-inflammatory, antibacterial and anticancer effects, its antimelanoma effect and underlying molecular mechanism have not been fully elucidated. In the current study, we investigated the inhibitory effect and action mechanism of BV and melittin against various melanoma cells including B16F10, A375SM and SK-MEL-28. BV and melittin potently suppressed the growth, clonogenic survival, migration and invasion of melanoma cells. They also reduced the melanin formation in α-melanocyte-stimulating hormone (MSH)-stimulated melanoma cells. Furthermore, BV and melittin induced the apoptosis of melanoma cells by enhancing the activities of caspase-3 and -9. In addition, we demonstrated that the antimelanoma effect of BV and melittin is associated with the downregulation of PI3K/AKT/mTOR and MAPK signaling pathways. We also found that the combination of melittin with the chemotherapeutic agent temozolomide (TMZ) significantly increases the inhibition of growth as well as invasion in melanoma cells compared to melittin or TMZ alone. Taken together, these results suggest that melittin could be potentially applied for the prevention and treatment of malignant melanoma.
Collapse
Affiliation(s)
- Haet Nim Lim
- Department of Pharmaceutical Engineering & Biotechnology, Sun Moon University, 70, Sunmoon-ro 221, Tangjeong-myeon, Asan-si, Chungnam 31460, Korea.
| | - Seung Bae Baek
- Eco system Lab., LOCORICO, Sun Moon University, 70, Sunmoon-ro 221, Tangjeong-myeon, Asan-si, Chungnam 31460, Korea.
| | - Hye Jin Jung
- Department of Pharmaceutical Engineering & Biotechnology, Sun Moon University, 70, Sunmoon-ro 221, Tangjeong-myeon, Asan-si, Chungnam 31460, Korea.
| |
Collapse
|
32
|
Antimutagenic and Synergistic Cytotoxic Effect of Cisplatin and Honey Bee Venom on 4T1 Invasive Mammary Carcinoma Cell Line. Adv Pharmacol Sci 2019; 2019:7581318. [PMID: 30838042 PMCID: PMC6374791 DOI: 10.1155/2019/7581318] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 12/09/2018] [Accepted: 12/13/2018] [Indexed: 11/17/2022] Open
Abstract
Introduction Honey bee venom (HBV) has various biological activities such as the inhibitory effect on several types of cancer. Cisplatin is an old and potent drug to treat most of the cancers. Our aim in the present study was to determine antimutagenic and cytotoxic effects of HBV on mammary carcinoma, exclusively and in combination with cisplatin. Methods In this study, 4T1 cell line was cultured in RPMI-1640 with 10% fetal bovine serum (FBS), at 37°C in humidified CO2 incubator. The cell viabilities were examined by the MTT assay. Also, HBV was screened for its antimutagenic activity via the Ames test. The results were assessed by SPSS software version 19 and one-way ANOVA method considering p < 0.05 level of significance. Results The results showed that 6 mg/ml of HBV, 20 μg/ml of cisplatin, and 6 mg/ml HBV with 10 μg/ml cisplatin could induce approximately 50% of 4T1 cell death. The concentration 7 mg/ml of HBV with of 62.76% inhibitory rate showed the highest antimutagenic activity in comparison with other treatment groups. Conclusions The MTT assay demonstrated that HBV and cisplatin could cause cell death in a dose-dependent manner. The cytotoxic effect of cisplatin also promoted by HBV. Ames test outcomes indicated that HBV could act as a significant mutagenic agent. The antimutagenic activity of HBV was increased considerably in the presence of S9 mix. Therefore, our findings have revealed that HBV can enhance the cytotoxic effect of cisplatin drug and its cancer-preventing effects.
Collapse
|
33
|
Rady I, Siddiqui IA, Rady M, Mukhtar H. Melittin, a major peptide component of bee venom, and its conjugates in cancer therapy. Cancer Lett 2017; 402:16-31. [PMID: 28536009 DOI: 10.1016/j.canlet.2017.05.010] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 02/07/2023]
Abstract
Melittin (MEL), a major peptide component of bee venom, is an attractive candidate for cancer therapy. This agent has shown a variety of anti-cancer effects in preclinical cell culture and animal model systems. Despite a convincing efficacy data against variety of cancers, its applicability to humans has met with challenges due to several issues including its non-specific cytotoxicity, degradation and hemolytic activity. Several optimization approaches including utilization of nanoparticle based delivery of MEL have been utilized to circumvent the issues. Here, we summarize the current understanding of the anticancer effects of bee venom and MEL on different kinds of cancers. Further, we also present the available information for the possible mechanism of action of bee venom and/or MEL.
Collapse
Affiliation(s)
- Islam Rady
- School of Medicine and Public Health, Department of Dermatology, University of Wisconsin-Madison, WI 53706, USA; Department of Zoology, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Imtiaz A Siddiqui
- School of Medicine and Public Health, Department of Dermatology, University of Wisconsin-Madison, WI 53706, USA
| | - Mohamad Rady
- Department of Zoology, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Hasan Mukhtar
- School of Medicine and Public Health, Department of Dermatology, University of Wisconsin-Madison, WI 53706, USA.
| |
Collapse
|
34
|
Wu Y, Han MF, Liu C, Liu TY, Feng YF, Zou Y, Li B, Liao HL. Design, synthesis, and antiproliferative activities of stapled melittin peptides. RSC Adv 2017. [DOI: 10.1039/c6ra26427a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Melittin is a 26-residue, amphipathic, cell-penetrating, α-helical anti-hepatoma peptide isolated from bee venom.
Collapse
Affiliation(s)
- Ye Wu
- School of Pharmacy
- Chengdu Medical College
- Chengdu 610083
- China
| | - Meng-fei Han
- Department of Traditional Chinese Medicine
- Changhai Hospital
- Second Military Medical University
- Shanghai 200433
- China
| | - Chao Liu
- Department of Organic Chemistry
- School of Pharmacy
- Second Military Medical University
- Shanghai 200433
- China
| | - Tai-yu Liu
- Department of Organic Chemistry
- School of Pharmacy
- Second Military Medical University
- Shanghai 200433
- China
| | - Yu-fei Feng
- Department of Organic Chemistry
- School of Pharmacy
- Second Military Medical University
- Shanghai 200433
- China
| | - Yan Zou
- Department of Organic Chemistry
- School of Pharmacy
- Second Military Medical University
- Shanghai 200433
- China
| | - Bai Li
- Department of Traditional Chinese Medicine
- Changhai Hospital
- Second Military Medical University
- Shanghai 200433
- China
| | - Hong-li Liao
- School of Pharmacy
- Chengdu Medical College
- Chengdu 610083
- China
| |
Collapse
|
35
|
Liu CC, Hao DJ, Zhang Q, An J, Zhao JJ, Chen B, Zhang LL, Yang H. Application of bee venom and its main constituent melittin for cancer treatment. Cancer Chemother Pharmacol 2016; 78:1113-1130. [DOI: 10.1007/s00280-016-3160-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/20/2016] [Indexed: 01/29/2023]
|
36
|
Béchohra L, Laraba-Djebari F, Hammoudi-Triki D. Cytotoxic activity of Androctonus australis hector venom and its toxic fractions on human lung cancer cell line. J Venom Anim Toxins Incl Trop Dis 2016; 22:29. [PMID: 27790250 PMCID: PMC5075196 DOI: 10.1186/s40409-016-0085-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/30/2016] [Indexed: 01/04/2023] Open
Abstract
Background Several studies have showed that animal venoms are a source of bioactive compounds that may inhibit the growth of cancer cells, which makes them useful agents for therapeutic applications. Recently, it was established that venom toxins from scorpions induced cytotoxic, antiproliferative and apoptogenic effects on cancer cells. Therefore, the present study aims to investigate the cytotoxic activity of Androctonus australis hector (Aah) scorpion venom and its toxic fractions (FtoxG-50 and F3) on NCI-H358 human lung cancer cells. Methods The cytotoxic and antiproliferative activities were estimated using MTT assay, lactate dehydrogenase release and clonogenic assays. Apoptosis was evaluated by Hoechst 33258 staining, DNA fragmentation assay and caspase-3 activity. Oxidative stress was analyzed by reactive oxygen species, nitric oxide, malondialdehyde and protein carbonyl levels along with assessment of antioxidant status. In addition, alteration of mitochondrial membrane potential was analyzed by JC1 fluorescent dye. Results The present findings showed that F3 fraction was more cytotoxic towards NCI-H358 lung cancer cells with an IC50 of 27.05 ± 0.70 μg/mL than venom alone (396.60 ± 1.33 μg/mL) and its toxic fraction FtoxG-50 (45.86 ± 0.91 μg/mL). Nevertheless, F3 fraction was not cytotoxic at these concentrations on normal human lung fibroblast MRC-5 cells. Inhibition of NCI-H358 cell proliferation after F3 fraction exposure occurred mainly by apoptosis as evidenced by damaged nuclei, significant DNA fragmentation level and caspase-3 activation in a dose dependent manner. Moreover, F3 fraction enhanced oxidative and nitrosative stress biomarkers and dissipated mitochondrial membrane potential in lung cancer cells along with significant depletion in cellular enzymatic and non-enzymatic antioxidants. Further, the apoptosis induced by F3 fraction was markedly prevented by the antioxidant N-acetylcysteine (NAC) suggesting the potential mechanism of oxidative stress. Conclusion These findings suggest that F3 fraction could induce apoptosis in lung cancer cells through involvement of oxidative stress and mitochondrial dysfunction. Hence, these properties make F3 fraction a promising candidate for development of new anticancer agents.
Collapse
Affiliation(s)
- Louisa Béchohra
- USTHB, Faculty of Biological Sciences, Laboratory of cellular and Molecular Biology, BP32, El Alia, Bab Ezzouar, 16111 Algiers, Algeria
| | - Fatima Laraba-Djebari
- USTHB, Faculty of Biological Sciences, Laboratory of cellular and Molecular Biology, BP32, El Alia, Bab Ezzouar, 16111 Algiers, Algeria
| | - Djelila Hammoudi-Triki
- USTHB, Faculty of Biological Sciences, Laboratory of cellular and Molecular Biology, BP32, El Alia, Bab Ezzouar, 16111 Algiers, Algeria
| |
Collapse
|
37
|
Gajski G, Čimbora-Zovko T, Rak S, Osmak M, Garaj-Vrhovac V. Antitumour action on human glioblastoma A1235 cells through cooperation of bee venom and cisplatin. Cytotechnology 2016; 68:1197-205. [PMID: 25916941 PMCID: PMC4960167 DOI: 10.1007/s10616-015-9879-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 04/15/2015] [Indexed: 01/06/2023] Open
Abstract
Cisplatin (cDDP) is one of the most widely used anticancer-drugs in both therapy and research. However, cDDP-resistance is the greatest obstacle for the successful treatment of cancer patients. In the present study, the possible joint anticancer effect of bee venom (BV), as a natural toxin, and cDDP towards human glioblastoma A1235 cells was evaluated. Treatment with BV alone in concentrations of 2.5-30 μg/ml displayed dose-dependent cytotoxicity towards A1235 cells, as evaluated with different cytotoxicity assays (MTT, Cristal violet and Trypan blue exclusion assay), with an IC50 value of 22.57 μg/ml based on the MTT results. Furthermore, BV treatment induced necrosis, which was confirmed by typical morphological features and fast staining with ethidium-bromide dye. Pre-treatment with BV induced cell sensitization to cDDP, indicating that BV could improve the killing effect of selected cells when combined with cDDP. The isobologram method used to determine the extent of synergism in combining two agents to examine their possible therapeutic effect showed that combined treatment induced an additive and/or synergistic effect towards selected cells depending on the concentration of both. Hence, a greater anticancer effect could be triggered if BV was used in the course of chemotherapy. The obtained results indicate that joint treatment with BV could be useful from the point of minimizing the cDDP concentration during chemotherapy, thus reducing and/or postponing the development of drug resistance. Our data, in accordance with previously reported results, suggests that BV could be used in the development of a new strategy for cancer treatment.
Collapse
Affiliation(s)
- Goran Gajski
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, 10000, Zagreb, Croatia
| | - Tamara Čimbora-Zovko
- Laboratory for Genotoxic Agents, Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000, Zagreb, Croatia
| | - Sanjica Rak
- Laboratory for Genotoxic Agents, Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000, Zagreb, Croatia
| | - Maja Osmak
- Laboratory for Genotoxic Agents, Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000, Zagreb, Croatia
| | - Vera Garaj-Vrhovac
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, 10000, Zagreb, Croatia.
| |
Collapse
|
38
|
Kong GM, Tao WH, Diao YL, Fang PH, Wang JJ, Bo P, Qian F. Melittin induces human gastric cancer cell apoptosis via activation of mitochondrial pathway. World J Gastroenterol 2016; 22:3186-3195. [PMID: 27003995 PMCID: PMC4789993 DOI: 10.3748/wjg.v22.i11.3186] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 11/17/2015] [Accepted: 12/12/2015] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the apoptotic effects of melittin on SGC-7901 cells via activation of the mitochondrial signaling pathway in vitro. METHODS SGC-7901 cells were stimulated by melittin, and its effect on proliferation and apoptosis of was investigated by methyl thiazolyl tetrazolium assay, morphologic structure with transmission electron microscopy, annexin-V/propidium iodide double-staining assay, measuring mitochondrial membrane potential (MMP) levels, and analyzing reactive oxygen species (ROS) concentrations were analyzed by flow cytometry. Cytochrome C (Cyt C), apoptosis-inducing factor (AIF), endonuclease G (Endo G), second mitochondria-derived activator of caspases (Smac)/direct IAP binding protein with low isoelectric point (Diablo), and FAS were analyzed by western blot. The expression of caspase-3 and caspase-8 was measured using activity assay kits. RESULTS Melittin was incubated at 1.0, 2.0, 4.0, or 6.0 μg/mL for 1, 2, 4, 6, or 8 h and showed a time- and concentration-dependent inhibition of SGC-7901 cell growth. Melittin induced SGC-7901 cell apoptosis, which was confirmed by typical morphological changes. Treatment with 4 μg/mL melittin induced early apoptosis of SGC-7901 cells, and the early apoptosis rates were 39.97% ± 3.19%, 59.27% ± 3.94%, and 71.50% ± 2.87% vs 32.63% ± 2.75% for 1, 2, and 4 h vs 0 h (n = 3, P < 0.05); the ROS levels were 616.53% ± 79.78%, 974.81% ± 102.40%, and 1330.94% ± 93.09% vs 603.74% ± 71.99% (n = 3, P < 0.05); the MMP values were 2.07 ± 0.05, 1.78 ± 0.29, and 1.16 ± 0.25 vs 2.55 ± 0.42 (n = 3, P < 0.05); caspase-3 activity was significantly higher compared to the control (5492.3 ± 321.1, 6562.0 ± 381.3, and 8695.7 ± 449.1 vs 2330.0 ± 121.9), but the caspase activity of the non-tumor cell line L-O2 was not different from that of the control. With the addition of the caspase-3 inhibitor (Ac-DEVD-CHO), caspase-3 activity was significantly decreased compared to the control group (1067.0 ± 132.5 U/g vs 8695.7 ± 449.1 U/g). The expression of the Cyt C, Endo G, and AIF proteins in SGC-7901 cells was significantly higher than those in the control (P < 0.05), while the expression of the Smac/Diablo protein was significantly lower than the control group after melittin exposure (P < 0.01). Ac-DEVD-CHO did not, however, have any effect on the expression of caspase-8 and FAS in the SGC-7901 cells. CONCLUSION Melittin can induce apoptosis of human gastric cancer (GC) cells through the mitochondria pathways, and it may be a potent agent in the treatment of human GC.
Collapse
|
39
|
Gajski G, Domijan AM, Žegura B, Štern A, Gerić M, Novak Jovanović I, Vrhovac I, Madunić J, Breljak D, Filipič M, Garaj-Vrhovac V. Melittin induced cytogenetic damage, oxidative stress and changes in gene expression in human peripheral blood lymphocytes. Toxicon 2016; 110:56-67. [DOI: 10.1016/j.toxicon.2015.12.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 12/07/2015] [Accepted: 12/11/2015] [Indexed: 12/12/2022]
|
40
|
|
41
|
Gajski G, Čimbora-Zovko T, Rak S, Rožman M, Osmak M, Garaj-Vrhovac V. Combined antitumor effects of bee venom and cisplatin on human cervical and laryngeal carcinoma cells and their drug resistant sublines. J Appl Toxicol 2015; 34:1332-41. [PMID: 25493319 DOI: 10.1002/jat.2959] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In the present study, we investigated the possible combined anticancer ability of bee venom (BV) and cisplatin towards two pairs of tumour cell lines: parental cervical carcinoma HeLa cells and their cisplatin-resistant HeLa CK subline,as well as laryngeal carcinoma HEp-2 cells and their cisplatin-resistant CK2 subline. Additionally, we identified several peptides of BV in the BV sample used in the course of the study and determined the exact concentration of MEL. BV applied alone in concentrations of 30 to 60 μg ml(–1) displayed dose-dependent cytotoxicity against all cell lines tested. Cisplatin-resistant cervical carcinoma cells were more sensitive to BV than their parental cell lines (IC(50) values were 52.50 μg ml(–1) for HeLa vs.47.64 μg ml(–1) for HeLa CK cells), whereas opposite results were obtained for cisplatin-resistant laryngeal carcinoma cells (IC(50) values were 51.98 μg ml(–1) for HEp-2 vs. > 60.00 μg ml(–1) for CK2 cells). Treatment with BV alone induced a necrotic type of cell death, as shown by characteristic morphological features, fast staining with ethidium-bromide and a lack of cleavage of apoptotic marker poly (ADP-ribose) polymerase (PARP) on Western blot. Combined treatment of BV and cisplatin induced an additive and/or weak synergistic effect towards tested cell lines, suggesting that BV could enhance the killing effect of selected cells when combined with cisplatin. Therefore, a greater anticancer effect could be triggered if BV was used in the course of chemotherapy. Our results suggest that combined treatment with BV could be useful from the point of minimizing the cisplatin concentration during chemotherapy, consequently reducing and/or postponing the development of cisplatin resistance.
Collapse
|
42
|
Khalil SR, Abd-Elhakim YM, Selim ME, Al-Ayadhi LY. Apitoxin protects rat pups brain from propionic acid-induced oxidative stress: The expression pattern of Bcl-2 and Caspase-3 apoptotic genes. Neurotoxicology 2015; 49:121-31. [PMID: 26048086 DOI: 10.1016/j.neuro.2015.05.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/27/2015] [Accepted: 05/31/2015] [Indexed: 10/23/2022]
Abstract
The primary aim of this study was to determine the potential modulatory role of the apitoxin (bee venom; BV) against propionic acid (PPA)-induced neurotoxicity. The biochemical responses to PPA exposure in rat pups were assayed, including changes in the antioxidant barrier systems and lipid peroxidation and protein oxidation biomarkers in the brain tissue. DNA damage was measured by single-cell gel electrophoresis and differences in Bcl-2 and Caspase-3 mRNA expression were assessed using real-time PCR. Changes in amygdala complex ultrastructure were visually assessed using electron microscopy. Sixty rat pups were assigned into six groups: a control group, a PPA-treated group, a BV-treated group, a protective co-treated group, a therapeutic co-treated group, and a protective/therapeutic co-treated group. The results indicate that PPA induced a pronounced increase (64.6%) in malondialdehyde (MDA), and in DNA damage (73.3%) with three-fold increase in protein carbonyl concentration. A significant reduction was observed in the enzyme activities of superoxide dismutase (SOD) (48.7%) and catalase (CAT) (74.8%) and reduced glutathione (GSH) level (52.6%). BV significantly neutralized the PPA-induced oxidative stress effects, especially in the BV protective/therapeutic co-treated group. In this group, GSH levels were restored to 64.5%, and MDA, protein carbonyl levels and tail moment % were diminished by 69.5, 21.1 and 18.8% relative to the control, respectively. Furthermore, while PPA induced significant apoptotic neural cell death, BV markedly inhibited apoptosis by promoting Bcl-2 expression and blocking Caspase-3 expression. BV markedly restored the normal ultrastructural morphology of the amygdala complex neurons. These results conclusively demonstrate that BV administration provides both protective and therapeutic effects in response to the PPA-induced deleterious effects, including oxidative stress, DNA damage, and neuronal death in the brains of rat pups.
Collapse
Affiliation(s)
- Samah R Khalil
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, Zagazig University, Egypt
| | - Yasmina M Abd-Elhakim
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, Zagazig University, Egypt
| | - Manar E Selim
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; Zoology Department, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | - Laila Y Al-Ayadhi
- Physiology Department, Faculty of Medicine, King Saud University, Autism Research and Treatment Centre, AL-Amodi Autism Research Chair, Riyadh, Saudi Arabia
| |
Collapse
|
43
|
Al-Ani I, Zimmermann S, Reichling J, Wink M. Pharmacological synergism of bee venom and melittin with antibiotics and plant secondary metabolites against multi-drug resistant microbial pathogens. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:245-55. [PMID: 25765829 DOI: 10.1016/j.phymed.2014.11.019] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 11/24/2014] [Accepted: 11/28/2014] [Indexed: 05/25/2023]
Abstract
The goal of this study was to investigate the antimicrobial activity of bee venom and its main component, melittin, alone or in two-drug and three-drug combinations with antibiotics (vancomycin, oxacillin, and amikacin) or antimicrobial plant secondary metabolites (carvacrol, benzyl isothiocyanate, the alkaloids sanguinarine and berberine) against drug-sensitive and antibiotic-resistant microbial pathogens. The secondary metabolites were selected corresponding to the molecular targets to which they are directed, being different from those of melittin and the antibiotics. The minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) were evaluated by the standard broth microdilution method, while synergistic or additive interactions were assessed by checkerboard dilution and time-kill curve assays. Bee venom and melittin exhibited a broad spectrum of antibacterial activity against 51 strains of both Gram-positive and Gram-negative bacteria with strong anti-MRSA and anti-VRE activity (MIC values between 6 and 800 µg/ml). Moreover, bee venom and melittin showed significant antifungal activity (MIC values between 30 and 100 µg/ml). Carvacrol displayed bactericidal activity, while BITC exhibited bacteriostatic activity against all MRSA and VRE strains tested (reference strains and clinical isolates), both compounds showed a remarkable fungicidal activity with minimum fungicidal concentration (MFC) values between 30 and 200 µg/ml. The DNA intercalating alkaloid sanguinarine showed bactericidal activity against MRSA NCTC 10442 (MBC 20 µg/ml), while berberine exhibited bacteriostatic activity against MRSA NCTC 10442 (MIC 40 µg/ml). Checkerboard dilution tests mostly revealed synergism of two-drug combinations against all the tested microorganisms with FIC indexes between 0.24 and 0.50, except for rapidly growing mycobacteria in which combinations exerted an additive effect (FICI = 0.75-1). In time-kill assays all three-drug combinations exhibited a powerful bactericidal synergistic effect against MRSA NCTC 10442, VRE ATCC 51299, and E. coli ATCC 25922 with a reduction of more than 3log10 in the colony count after 24 h. Our findings suggest that bee venom and melittin synergistically enhanced the bactericidal effect of several antimicrobial agents when applied in combination especially when the drugs affect several and differing molecular targets. These results could lead to the development of novel or complementary antibacterial drugs against MDR pathogens.
Collapse
Affiliation(s)
- Issam Al-Ani
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, INF 364 Heidelberg, Germany; Department of Medical Laboratory Technology, Faculty of Medical Technology, Baghdad, Iraq
| | - Stefan Zimmermann
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University, INF 324 Heidelberg, Germany
| | - Jürgen Reichling
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, INF 364 Heidelberg, Germany
| | - Michael Wink
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, INF 364 Heidelberg, Germany.
| |
Collapse
|
44
|
Behroozi J, Divsalar A, Saboury AA. Honey bee venom decreases the complications of diabetes by preventing hemoglobin glycation. J Mol Liq 2014. [DOI: 10.1016/j.molliq.2014.09.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
45
|
Forsythiaside Protects Against Hydrogen Peroxide-Induced Oxidative Stress and Apoptosis in PC12 Cell. Neurochem Res 2014; 40:27-35. [DOI: 10.1007/s11064-014-1461-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 10/16/2014] [Accepted: 10/18/2014] [Indexed: 12/20/2022]
|
46
|
Abstract
Melittin is a major active component of bee venom, and it exhibits strong biological activity. Recent reports have demonstrated that melittin has anti-tumor effects on many kinds of tumor cells through direct interaction with cell transduction mediators and influencing cellular signaling pathways, induction of apoptotic or necrotic cell death, and inhibition of growth and proliferationin vivo or in vitro. Nanotechnology and genetic engineering provide technical support to the safe and targeted delivery of melittin to the body. This review summarizes recent findings on the anti-tumor potential of melittin in tumors of different types.
Collapse
|
47
|
Combined cytogenotoxic effects of bee venom and bleomycin on rat lymphocytes: an in vitro study. BIOMED RESEARCH INTERNATIONAL 2014; 2014:173903. [PMID: 24822179 PMCID: PMC4009237 DOI: 10.1155/2014/173903] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/21/2014] [Indexed: 01/24/2023]
Abstract
This study was carried out to determine the cytotoxic and genotoxic effects of bee venom (BV) and/or the chemotherapeutic agent bleomycin (BLM) on healthy isolated rat lymphocytes utilizing morphometric and molecular techniques. Using the Ficoll-Histopaque density gradient centrifugation technique, lymphocytes were isolated, divided into groups, and subjected to BV and/or BLM at incubation medium concentrations of 10 or 20 μg/mL respectively for 24 and 72 hrs. An MTT assay and fluorescent microscopy examinations were used to assess the cytotoxic effects. To determine the predominant type of BV and/or BLM-induced cell death, LDH release assay was employed beside quantitative expression analyses of the apoptosis-related genes (Caspase-3 and Bcl-2). The genotoxic effects of the tested compounds were evaluated via DNA fragmentation assay. The results of these assays demonstrated that BV potentiates BLM-induced cytotoxicity through increased LDH release and diminished cell viability. Nevertheless, BV significantly inhibited the BLM-induced DNA damage. The results verify that BV significantly attenuates the genotoxic effects of BLM on noncancerous isolated rat lymphocytes but does not diminish BLM cytotoxicity.
Collapse
|
48
|
Hoshina MM, Marin-Morales MA. Anti-genotoxicity and anti-mutagenicity of Apis mellifera venom. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2014; 762:43-8. [PMID: 24525379 DOI: 10.1016/j.mrgentox.2013.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 08/16/2013] [Accepted: 11/02/2013] [Indexed: 11/30/2022]
Abstract
The search for substances able to inhibit and/or diminish the effects of genotoxic and mutagenic substances has been the target of several investigations performed in recent times. Hymenoptera venoms constitute a considerable source of substances with pharmacological potential. The present study aimed to evaluate the cytotoxic, genotoxic and anti-genotoxic, mutagenic and anti-mutagenic potentials of Apis mellifera venom in HepG2 cells. In this evaluation, the MTT test was applied to determine the most appropriate concentrations for the genotoxicity and mutagenicity tests. It was verified that the concentrations of 0.1, 0.05 and 0.01μg/mL were not cytotoxic, hence these concentrations were used in the experiments. For the evaluation of the genotoxic and mutagenic potential of the bee venom the comet assay and the micronucleus test were applied, respectively. The concentrations mentioned above presented both genotoxic and mutagenic potential for HepG2 cells and it was necessary to test lower concentrations of the venom (10pg/mL, 1pg/mL and 0.1pg/mL) for the anti-genotoxicity and anti-mutagenicity tests, which were performed subjecting the cells to the action of MMS (methyl methanesulfonate) in order to verify the ability of the venom to inhibit or diminish the action of this compound, which has a recognized action on the genetic material. Pre-, post-treatment and simultaneous treatment with and without incubation with the venom were performed. It was observed that the lowest three concentrations tested did not present any anti-genotoxic and anti-mutagenic activity on the cells. The use of bee venom for pharmacological purposes in treatments such as cancer must be done with extreme caution, since it was observed that even at very low concentrations the venom can induce genotoxicity and mutagenicity in human cells, as was verified for the HepG2 cells.
Collapse
Affiliation(s)
- Márcia Miyuki Hoshina
- Departamento de Biologia, Instituto de Biociências, UNESP, Avenida 24 A, 1515, CP 199, 13506-900 Rio Claro, SP, Brazil
| | - Maria Aparecida Marin-Morales
- Departamento de Biologia, Instituto de Biociências, UNESP, Avenida 24 A, 1515, CP 199, 13506-900 Rio Claro, SP, Brazil.
| |
Collapse
|
49
|
Safaeinejad Z, Nabiuni M, Nazari Z. Potentiation of a novel palladium (II) complex lethality with bee venom on the human T-cell acute lymphoblastic leukemia cell line (MOLT-4). J Venom Anim Toxins Incl Trop Dis 2013; 19:25. [PMID: 24090289 PMCID: PMC4015889 DOI: 10.1186/1678-9199-19-25] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 09/03/2013] [Indexed: 01/28/2023] Open
Abstract
Background Although honeybee venom (BV) has been reported to induce apoptosis in different types of cancerous cells, its synergistic effects with customary anti-cancer drugs remain largely unknown. In the present study, we evaluated the cytotoxic effect of BV alone (as a natural product) and the synergistic cytological effects of this component in combination with [Pd (bpy) (Pi-Pydtc)]NO3 – a novel palladium complex on human T-cell lymphoblastic leukemia cells. To investigate the cytotoxic effect of the BV alone and in combination with palladium complex on MOLT-4 cells MTT assay was performed. In order to determine the apoptotic effects of BV separately and in combination with Pd (II) complex on these cells and its ability to induce apoptosis, morphological examination, flowcytometric analysis and caspase-3 colorimetric assay were done. Results We found that BV induced morphological changes, namely nuclear shrinkage, and inhibited MOLT-4 cell proliferation; both effects were dose- and time-dependent. Flow cytometry by Annexin-V antibody demonstrated that BV induced apoptosis in MOLT-4 cells. Furthermore, BV induced apoptosis independently of caspase-3 in these cells. In addition, we proved a clear synergistic effect of BV on [Pd (bpy) (Pi-Pydtc)]NO3. The apoptotic pathway activated by BV in combination with Pd complex was caspase-3-dependent. Conclusions These observations provide an explanation for the anti-proliferative properties of BV, and suggest that this agent may be useful for treating lymphoblastic leukemia alone or in combination with chemotherapy drugs pending further investigations on animal models as preclinical tests.
Collapse
Affiliation(s)
- Zahra Safaeinejad
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | | | | |
Collapse
|
50
|
Kim SH, Jung SY, Lee KW, Lee SH, Cai M, Choi SM, Yang EJ. Bee venom effects on ubiquitin proteasome system in hSOD1(G85R)-expressing NSC34 motor neuron cells. Altern Ther Health Med 2013; 13:179. [PMID: 23866691 PMCID: PMC3722004 DOI: 10.1186/1472-6882-13-179] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 07/11/2013] [Indexed: 12/02/2022]
Abstract
Background Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that results from a progressive loss of motor neurons. Familial ALS (fALS) is caused by missense mutations in Cu, Zn-superoxide dismutase 1 (SOD1) that frequently result in the accumulation of mutant protein aggregates that are associated with impairments in the ubiquitin-proteasome system (UPS). UPS impairment has been implicated in many neurological disorders. Bee venom (BV) extracted from honey bees has been used as a traditional medicine for treating inflammatory diseases and has been shown to attenuate the neuroinflammatory events that occur in a symptomatic ALS animal model. Methods NSC34 cells were transiently transfected with a WT or G85R hSOD1-GFP construct for 24 hrs and then stimulated with 2.5 μg/ml BV for 24 hrs. To determine whether a SOD1 mutation affects UPS function in NSC34 cells, we examined proteasome activity and performed western blotting and immunofluorescence using specific antibodies, such as anti-misfolded SOD1, anti-ubiquitin, anti-GRP78, anti-LC3, and anti-ISG15 antibodies. Results We found that GFP-hSOD1G85R overexpression induced SOD1 inclusions and reduced proteasome activity compared with the overexpression of GFP alone in NSC34 motor neuronal cells. In addition, we also observed that BV treatment restored proteasome activity and reduced the accumulation of ubiquitinated and misfolded SOD1 in GFP-hSOD1G85R-overexpressing NSC34 motor neuronal cells. However, BV treatment did not activate the autophagic pathway in these cells. Conclusion Our findings suggest that BV may rescue the impairment of the UPS in ALS models.
Collapse
|