Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 316
Filtrar
1.
Dent Med Probl ; 61(1): 53-64, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38441304

RESUMO

BACKGROUND: Diabetes mellitus (DM) is a critical chronic metabolic disease. Several treatment modalities are currently under investigation. Both bee venom (BV) and bone marrow mesenchymal stem cells (BMSCs) can possibly offer an approach for treating type I diabetes. OBJECTIVES: The aim of the present study was to investigate the mechanism underlying the anti-diabetic effect of BV as compared to BMSCs on the tongue mucosa of diabetic rats. MATERIAL AND METHODS: A total of 52 male albino rats were used in the current study. The rats were randomly assigned into 4 groups: group 1 (control); group 2 (streptozocin (STZ)); group 3 (BV-treated); and group 4 (BMSC-treated). Diabetes mellitus was induced via an intraperitoneal (IP) injection of STZ in the rats from groups 2, 3 and 4. Following the diagnosis of DM, the rats in group 3 were injected with a daily dose of 0.5 mg/kg of BV, while the rats in group 4 were treated with a single injection of BMSCs. All rats were euthanized after 4 weeks, and their tongues were dissected and divided into halves. The right halves of the tongues were utilized for the histological examination, followed by morphometric analysis. In contrast, the left halves were used to detect the local gene expression of transforming growth factor beta 1 (TGF-ß1) and vascular endothelial growth factor (VEGF). RESULTS: Group 2 revealed marked disruption in the morphology of the fungiform and filiform papillae, and atrophic epithelial changes in both dorsal and ventral surface epithelium as compared to other groups. Group 4 showed a significantly larger number of taste buds, and a higher gene expression of TGF-ß1 and VEGF as compared to groups 2 and 3. Additionally, BV and BMSCs effectively increased the thickness of dorsal and ventral surface epithelium as compared to group 2. CONCLUSIONS: Treatment with BMSCs was associated with significant improvement in the morphology and number of lingual epithelial cells and taste buds in the tongues of diabetic rats as compared to BV-treated rats, which was due to the local upregulation of TGF-ß1 and VEGF gene expression.


Assuntos
Venenos de Abelha , Diabetes Mellitus Experimental , Células-Tronco Mesenquimais , Masculino , Animais , Ratos , Fator de Crescimento Transformador beta1 , Fator A de Crescimento do Endotélio Vascular , Diabetes Mellitus Experimental/terapia , Língua , Venenos de Abelha/farmacologia
2.
Front Immunol ; 15: 1326033, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38318188

RESUMO

Melittin, a main component of bee venom, is a cationic amphiphilic peptide with a linear α-helix structure. It has been reported that melittin can exert pharmacological effects, such as antitumor, antiviral and anti-inflammatory effects in vitro and in vivo. In particular, melittin may be beneficial for the treatment of diseases for which no specific clinical therapeutic agents exist. Melittin can effectively enhance the therapeutic properties of some first-line drugs. Elucidating the mechanism underlying melittin-mediated biological function can provide valuable insights for the application of melittin in disease intervention. However, in melittin, the positively charged amino acids enables it to directly punching holes in cell membranes. The hemolysis in red cells and the cytotoxicity triggered by melittin limit its applications. Melittin-based nanomodification, immuno-conjugation, structural regulation and gene technology strategies have been demonstrated to enhance the specificity, reduce the cytotoxicity and limit the off-target cytolysis of melittin, which suggests the potential of melittin to be used clinically. This article summarizes research progress on antiviral, antitumor and anti-inflammatory properties of melittin, and discusses the strategies of melittin-modification for its future potential clinical applications in preventing drug resistance, enhancing the selectivity to target cells and alleviating cytotoxic effects to normal cells.


Assuntos
Venenos de Abelha , Meliteno , Meliteno/farmacologia , Meliteno/química , Meliteno/metabolismo , Peptídeos Antimicrobianos , Venenos de Abelha/farmacologia , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Antivirais
3.
Arch Pharm (Weinheim) ; 357(4): e2300569, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38251938

RESUMO

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.


Assuntos
Venenos de Abelha , Neoplasias , Humanos , Meliteno/farmacologia , Meliteno/química , Meliteno/metabolismo , Relação Estrutura-Atividade , Venenos de Abelha/farmacologia , Venenos de Abelha/uso terapêutico , Neoplasias/tratamento farmacológico , Peptídeos/química
4.
Sci Rep ; 14(1): 1510, 2024 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-38233443

RESUMO

Breast cancer ranks as the second leading most significant of mortality for women. Studies have demonstrated the potential benefits of natural compounds in cancer treatment and prevention, either in isolation or in conjunction with chemotherapy. In order to improve Tamoxifen's therapeutic efficacy in in-vivo studies, our research sought to determine the effects of hesperidin, piperine, and bee venom as natural compounds, as well as their combination effect with or without Tamoxifen. First, 132 female albino rats were equally divided into six groups and five subgroups, and breast cancer was induced in the selected groups by xenografting of MCF7 cells. Second, the effect of single and best ratio combinations treatment from previous in vitro studies were selected. Next, tumorous mammary glands were collected for apoptotic and antiapoptotic biomarkers and cell cycle analysis. Single or combined natural products with or without Tamoxifen revealed a significant up-regulation in apoptotic genes Bax and Casp3 and a downregulation of antiapoptotic and angiogenesis genes Bcl-2 and VEGF genes. We found that cell cycle arrest in the G0/G1 phase was exclusively caused by Tamoxifen and/ or hesperidin. However, the cell cycle arrest in the G2/M phase is a result of the combination of piperine and bee venom, with or without Tamoxifen by using the flow cytometric technique. Our research concludes that bee venom, hesperidin, and piperine can synergistically enhance to increase Tamoxifen's efficiency in the management of breast cancer.


Assuntos
Alcaloides , Venenos de Abelha , Benzodioxóis , Neoplasias da Mama , Hesperidina , Piperidinas , Alcamidas Poli-Insaturadas , Humanos , Feminino , Ratos , Animais , Tamoxifeno/farmacologia , Tamoxifeno/uso terapêutico , Hesperidina/farmacologia , Hesperidina/uso terapêutico , Células MCF-7 , Venenos de Abelha/farmacologia , Venenos de Abelha/uso terapêutico , Angiogênese , Apoptose , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Biomarcadores
5.
Mol Biol Rep ; 51(1): 2, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-38057592

RESUMO

BACKGROUND: Lung cancer, one of the most common oncological diseases worldwide, continues to be the leading cause of cancer-related deaths. The development of new approaches for lung cancer, which still has a low survival rate despite medical advances, is of great importance. METHODS AND RESULTS: In this study, bee venom (BV), conditioned medium of MSCs isolated from dental follicles (MSC-CM) and cisplatin were applied at different doses and their effects on A549 cell line were evaluated. Dental follicles were used as a source of MSCs source and differentiation kits, and characterization studies (flow cytometry) were performed. Cell viability was measured by the MTT method and apoptosis was measured by an Annexin V-FITC/PI kit on flow cytometer. IC50 dose values were determined according to the 24th hour and were determined as 15.8 µg/mL for BV, 10.78% for MSC-CM and 5.77 µg/mL for cisplatin. IC50 values found for BV and MSC-CM were also given in combination and the effects were observed. It was found that the applied substances caused BV to decrease in cell viability and induced apoptosis in cells. In addition to the induction of apoptosis in BV, MSC-CM, and combined use, all three applications led to an increase in Bax protein expression and a decrease in Bcl-2 protein expression. The molecular mechanism of anticancer activity through inhibition of Bax and Bcl-2 proteins and the NF-κB signaling pathway may be suggested. CONCLUSION: Isolated MSCs in our study showed anticancer activity and BV and MSC-CM showed synergistic antiproliferative and apoptotic effects.


Assuntos
Venenos de Abelha , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Células-Tronco Mesenquimais , Humanos , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Cisplatino/farmacologia , Cisplatino/metabolismo , Neoplasias Pulmonares/metabolismo , Venenos de Abelha/farmacologia , Venenos de Abelha/metabolismo , Apoptose , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células-Tronco Mesenquimais/metabolismo
6.
Res Vet Sci ; 165: 105050, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37856942

RESUMO

Honey bee venom is a valuable product with a wide range of biological effects, whose use is rapidly increasing in apitherapy. In this study, the effect of gamma-irradiated honey bee venom (doses of 0, 2, 4, 6, and 8 kGy, volume of 0.1 ml, and concentration of 0.2 mg/ml) was evaluated on median lethal dose (LD50) determinations, liver and kidney histology, biochemical marker level, and serum protein analyses. Hence, the LD50 induced by the honey bee venom irradiated at 4, 6, and 8 kGy was increased, compared with the one at 0 and 2 kGy. Normal histology was observed in the liver and kidney of the mice receiving the honey bee venom irradiated at 4, 6, and 8 kGy. The serum levels of alanine aminotransferase (ALT) and all serum proteins were reduced at 4, 6, and 8 kGy compared with 0 and 2 kGy. Therefore, gamma irradiation at 4, 6, and 8 kGy had no negative effect on LD50, liver and kidney tissues, ALT, and serum protein levels by decreasing the allergen compounds of the honey bee venom.


Assuntos
Venenos de Abelha , Camundongos , Animais , Venenos de Abelha/farmacologia , Fígado , Alérgenos , Rim , Modelos Animais , Proteínas Sanguíneas
7.
Environ Sci Pollut Res Int ; 30(38): 88685-88703, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37442924

RESUMO

This study investigated the possible beneficial role of the bee venom (BV, Apis mellifera L.) against zinc oxide nanoparticles (ZNPs)-induced neurobehavioral and neurotoxic impacts in rats. Fifty male Sprague Dawley rats were alienated into five groups. Three groups were intraperitoneally injected distilled water (C 28D group), ZNPs (100 mg/kg b.wt) (ZNPs group), or ZNPs (100 mg/kg.wt) and BV (1 mg/ kg.bwt) (ZNPs + BV group) for 28 days. One group was intraperitoneally injected with 1 mL of distilled water for 56 days (C 56D group). The last group was intraperitoneally injected with ZNPs for 28 days, then BV for another 28 days at the same earlier doses and duration (ZNPs/BV group). Depression, anxiety, locomotor activity, spatial learning, and memory were evaluated using the forced swimming test, elevated plus maze, open field test, and Morris water maze test, respectively. The brain contents of dopamine, serotonin, total antioxidant capacity (TAC), malondialdehyde (MDA), and Zn were estimated. The histopathological changes and immunoexpressions of neurofilament and GAP-43 protein in the brain tissues were followed. The results displayed that BV significantly decreased the ZNPs-induced depression, anxiety, memory impairment, and spatial learning disorders. Moreover, the ZNPs-induced increment in serotonin and dopamine levels and Zn content was significantly suppressed by BV. Besides, BV significantly restored the depleted TAC but minimized the augmented MDA brain content associated with ZNPs exposure. Likewise, the neurodegenerative changes induced by ZNPs were significantly abolished by BV. Also, the increased neurofilament and GAP-43 immunoexpression due to ZNPs exposure were alleviated with BV. Of note, BV achieved better results in the ZNPs + BV group than in the ZNPs/BV group. Conclusively, these results demonstrated that BV could be employed as a biologically effective therapy to mitigate the neurotoxic and neurobehavioral effects of ZNPs, particularly when used during ZNPs exposure.


Assuntos
Venenos de Abelha , Nanopartículas , Síndromes Neurotóxicas , Óxido de Zinco , Ratos , Animais , Masculino , Abelhas , Ratos Sprague-Dawley , Proteína GAP-43/metabolismo , Proteína GAP-43/farmacologia , Óxido de Zinco/metabolismo , Venenos de Abelha/farmacologia , Venenos de Abelha/toxicidade , Dopamina/metabolismo , Dopamina/farmacologia , Serotonina/metabolismo , Filamentos Intermediários/metabolismo , Antioxidantes/metabolismo , Síndromes Neurotóxicas/metabolismo , Encéfalo
8.
Nutrients ; 15(14)2023 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-37513529

RESUMO

Apitherapy (using bee products) has gained broad recognition in cancer therapeutics globally. Honeybee venom has a broad range of biological potential, and its utilization is rapidly emerging in apitherapy. Bee products have significant potential to strengthen the immune system and improve human health. Thus, this review is targeted toward recapitulating the chemo-preventive potential of melittin (MEL), which constitutes a substantial portion of honeybee venom. Honeybee venom (apitoxin) is produced in the venom gland of the honeybee abdomen, and adult bees utilize it as a primary colony defense mechanism. Apitoxin comprises numerous biologically active compounds, including peptides, enzymes, amines, amino acids, phospholipids, minerals, carbohydrates, and volatile components. We are mainly focused on exploring the potential of melittin (a peptide component) of bee venom that has shown promising potential in the treatment of several human cancers, including breast, stomach, lung, prostate, ovary, kidney, colon, gastric, esophageal, cervical cancers, melanoma, osteosarcoma, and hepatocellular carcinoma. This review has summarized all potential studies related to the anticancerous efficacy of melittin (apitoxin), its formulations, conjugates, and nano-formulations against several human carcinomas, which would further pave the way for future researchers in developing potent drugs for cancer management.


Assuntos
Venenos de Abelha , Neoplasias Ósseas , Carcinoma Hepatocelular , Neoplasias Hepáticas , Masculino , Humanos , Abelhas , Animais , Venenos de Abelha/farmacologia , Venenos de Abelha/uso terapêutico , Meliteno/farmacologia , Meliteno/uso terapêutico , Peptídeos
9.
Chem Biodivers ; 20(6): e202300401, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37227219

RESUMO

Bee venom is a natural mixture and candidate anti-cancer agent with selective cytotoxic effect on some cancer cells. However, the cellular mechanisms of how bee venom selectively targets cancer cells remain elusive. The aim of this study was to reveal the genotoxic effect of bee venom in concordance with the location of ß-actin protein throughout the nucleus or/and cytoplasm. For this aim, the level of H2AX phosphorylation (γH2AX) and intracellular location of ß-actin were assessed by immunofluorescence in liver (HEPG2) and metastatic breast (MDA-MB-231) cancer cell lines compared to normal fibroblasts (NIH3T3) after bee venom treatment. Colocalisation profiles of γH2AX and ß-actin in each cell line were also analysed. The results showed that the levels of γH2AX staining decreased in normal cells but increased in cancer cells. The majority of ß-actin was localised within the cytoplasm of normal cells after bee venom treatment, but it was mostly accumulated within the nucleus in cancer cells. Colocalisation of ß-actin and γH2AX both in nucleus and cytoplasm was induced in each cancer cell by different patterns. The results showed that normal and cancerous cells had different responses against bee venom, and suggested that bee venom induced a cellular response by the interaction between γH2AX and ß-actin.


Assuntos
Venenos de Abelha , Neoplasias , Animais , Camundongos , Venenos de Abelha/farmacologia , Histonas/metabolismo , Actinas/metabolismo , Células NIH 3T3 , Fígado/metabolismo
10.
Biomolecules ; 13(4)2023 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-37189427

RESUMO

Peripheral nerve injury (PNI) is a health problem that affects many people worldwide. This study is the first to evaluate the potential effect of bee venom (BV) and its major components in a model of PNI in the mouse. For that, the BV used in this study was analyzed using UHPLC. All animals underwent a distal section-suture of facial nerve branches, and they were randomly divided into five groups. Group 1: injured facial nerve branches without any treatment. Group 2: the facial nerve branches were injured, and the normal saline was injected similarly as in the BV-treated group. Group 3: injured facial nerve branches with local injections of BV solution. Group 4: injured facial nerve branches with local injections of a mixture of PLA2 and melittin. Group 5: injured facial nerve branches with local injection of betamethasone. The treatment was performed three times a week for 4 weeks. The animals were submitted to functional analysis (observation of whisker movement and quantification of nasal deviation). The vibrissae muscle re-innervation was evaluated by retrograde labeling of facial motoneurons in all experimental groups. UHPLC data showed 76.90 ± 0.13%, 11.73 ± 0.13%, and 2.01 ± 0.01%, respectively, for melittin, phospholipase A2, and apamin in the studied BV sample. The obtained results showed that BV treatment was more potent than the mixture of PLA2 and melittin or betamethasone in behavioral recovery. The whisker movement occurred faster in BV-treated mice than in the other groups, with a complete disappearance of nasal deviation two weeks after surgery. Morphologically, a normal fluorogold labeling of the facial motoneurons was restored 4 weeks after surgery in the BV-treated group, but no such restoration was ever observed in other groups. Our findings indicate the potential of the use of BV injections to enhance appropriate functional and neuronal outcomes after PNI.


Assuntos
Venenos de Abelha , Traumatismos do Nervo Facial , Animais , Camundongos , Venenos de Abelha/farmacologia , Venenos de Abelha/uso terapêutico , Betametasona , Traumatismos do Nervo Facial/tratamento farmacológico , Meliteno/farmacologia , Meliteno/uso terapêutico , Fosfolipases A2
11.
BMC Complement Med Ther ; 23(1): 132, 2023 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-37098530

RESUMO

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.


Assuntos
Venenos de Abelha , Leucemia , Humanos , Abelhas , Animais , Meliteno/farmacologia , Meliteno/química , Meliteno/genética , Venenos de Abelha/farmacologia , Células K562 , Peptídeos , Leucemia/tratamento farmacológico
12.
Sci Rep ; 13(1): 6436, 2023 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-37081055

RESUMO

The misuse of antibiotics has led to antibiotic-resistant bacterial strains, making it even harder to combat and eliminate their infections. Staphylococcus aureus causes various adverse infections and diseases, including skin abscesses, bloodstream infections, pneumonia, and joint infections. In this study, we aimed to test the cytotoxic and antibacterial effects of bee venom-loaded chitosan nanoparticles (BV-loaded CS-NPs) in comparison to gamma-irradiated BV and native BV from Apis mellifera. The physiochemical characterizations of our treatments were determined by Fourier Transform Infrared Spectroscopy (FTIR), Transmission Electron Microscope (TEM), zeta-potential, release rate, and Encapsulation Efficiency (EE). Our study was conducted on both levels, in-vitro and in-vivo. For the in-vitro study, a bacterial model of Staphylococcus aureus with an ATCC number of 6538 was grown in tryptic soy agar (TSA) medium, and the inhibition zones of our drug candidates were measured with the appropriate statistical analysis performed. For the in-vivo study, levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), Creatinine, Urea, and interleukin 6 (IL-6) were analyzed. BV-loaded CS-NPs showed relatively better results than the other alternatives, which are native BV and gamma-irradiated BV. The results showed that the antibacterial effect of BV-loaded CS-NPs was greater than the alternatives. Furthermore, its cytotoxic effect was far less than the native and irradiated bee venom. These outcomes ensure that loading BV on CS-NPs makes it a promising drug candidate for an antibiotic alternative with minimal cytotoxicity and enhanced antibacterial activity.


Assuntos
Antineoplásicos , Venenos de Abelha , Quitosana , Nanopartículas , Infecções Estafilocócicas , Abelhas , Animais , Antibacterianos/farmacologia , Antibacterianos/química , Staphylococcus aureus , Venenos de Abelha/farmacologia , Infecções Estafilocócicas/tratamento farmacológico , Nanopartículas/química , Antineoplásicos/farmacologia , Quitosana/farmacologia , Quitosana/química
13.
Egypt J Immunol ; 30(2): 11-25, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37031394

RESUMO

Bee venom (BV) showed therapeutical effects to treat various diseases as it contains at least 18 pharmacologically active components including various enzymes, peptides, and amines. This study aimed to evaluate the action of BV on some hematological parameters, humoral and cellular immunity, and the determination of antioxidant levels in male albino rats. The study included 40 male albino rats (190-210 g), divided into four groups. Three groups were injected subcutaneously with three different doses of BV (2.5, 5, and 10 mg/kg, respectively). The control group was injected with saline solution. Blood samples were obtained to measure total leucocytes count (TLC), differential leukocytes count, hematological parameters (hemoglobin (Hb), hematocrit (HCT), red blood cells (RBCs), mean cell volume (MCV), mean cell hemoglobin (MCH), mean cell hemoglobin concentration (MCHC) and Platelets. Sera were used to assess immunoglobulins (IgM, IgG, IgA, and IgE), some cytokines e.g., tumor necrosis factor-alpha (TNF-α), tumor growth factor beta (TGF-ß), interleukins 6 and 10 (IL-6, IL-10), and some antioxidant levels malondialdehyde (MDA), super oxide dismutase (SOD), and glutathione (GSH). Data showed that BV therapy increased antibody production levels (IgM, IgG, and IgA) while decreasing IgE levels. Hematological markers (Hb and lymphocytes) were increased. BV increased total TGF- ß and IL-10 but decreased total TNF- α and IL-6. On the antioxidant scale, an increase in SOD, CAT, and GSH levels was observed, accompanied by a decrease in MDA levels. However, the BV treatment led to a significant reduction in the number of eosinophils, monocytes, and neutrophils (p < 0.05). In conclusion, our findings suggested that BV may be utilized to increase the effectiveness of various immunological and hematological parameters.


Assuntos
Antioxidantes , Venenos de Abelha , Masculino , Humanos , Ratos , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Interleucina-10 , Interleucina-6 , Venenos de Abelha/farmacologia , Superóxido Dismutase , Fator de Necrose Tumoral alfa , Hemoglobinas , Imunoglobulina A , Imunoglobulina E , Imunoglobulina G , Imunoglobulina M , Animais
14.
Biomed Pharmacother ; 161: 114521, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36921536

RESUMO

Methylmercury (MeHg) toxicity is associated with extensive neuronal degeneration of dorsal root ganglia (DRG). This study aimed to assess the ameliorative effect of bee venom (BV) on methyl mercury chloride (MeHgCl)-induced peripheral neurotoxicity using DRGs in rats. Forty-eight adult male Sprague Dawley rats were allocated into four equal groups: G I: control (gavaged MilliQ water 1 ml/rat), G II: subcutaneously injected with BV (0.5 mg/kg b.wt), G III: gavaged MeHgCl (6.7 mg/kg b.wt), and G IV: received MeHgCl+BV. Dosing was done five times/week for 2 weeks. Ataxic behavior and visual impairments were significantly increased, whereas the movement behavior and motility gait were suppressed in the MeHgCl group. MeHgCl significantly decreased total antioxidant capacity (TAC) in DRG and significantly decreased the serum levels of glutathione (GSH), catalase (CAT), and superoxide dismutase (SOD). Tumor necrosis factor-alpha (TNF-α) and interleukin 1ß (IL-1ß) levels were significantly elevated, whereas interleukin 10 (IL-10) levels were significantly decreased in the MeHgCl group compared with the control group. DRGs of the MeHgCl-exposed rats showed pyknotic shrunken neurons with perineural vacuolations, demyelination of nerve axons, and proliferation of the satellite cells. MeHgCl significantly induced a higher positive index ratio of Iba-1, SOX10, neurofilament, pan-neuron, and vimentin immunostaining in the DRG. BV administration significantly mitigated the MeHgCl-induced alterations in oxidative stress-related indices. BV modified the immunostaining of Iba-1, SOX10, neurofilament, pan-neuron, and vimentin-positive index ratio in the DRG of the MeHgCl group. Our findings acknowledged that BV could enhance in vivo neuroprotective effects against MeHgCl-induced DRGs damage in male rats.


Assuntos
Venenos de Abelha , Mercúrio , Compostos de Metilmercúrio , Ratos , Animais , Masculino , Compostos de Metilmercúrio/toxicidade , Ratos Sprague-Dawley , Vimentina , Gânglios Espinais , Venenos de Abelha/farmacologia , Estresse Oxidativo , Glutationa/farmacologia
15.
Chem Biodivers ; 20(3): e202201263, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36806913

RESUMO

Honeybees provide multiple products such as bee venom (BV) which are used for various nutritional and medicinal purposes. BV has received great attention due to its wide range of bioactive components with potential anti-cancer effects on different cancers. Triple negative breast cancer (TNBC) is defined as an aggressive type of breast cancer and new therapeutic targets are required for its treatment. In the current literature information is varied about the composition and quantity of BV bioactive compounds as well as the origin of BV and its significance. In this context, the cytotoxic and apoptotic effects of BV with a higher rate of mellitin from Apis mellifera anatoliaca (Mugla ecotype) on MDA-MB-231 cells was evaluated, in vitro. The cytotoxic, apoptotic and morphological effects of BV were determined by WST-1, Annexin V, cell cycle analysis and Acridine Orange staining. The results showed that BV caused apoptotic cell death in TNBC cells at a lower dose (0.47 µg/mL, p<0.01). This study suggests that BV could be developed as a potential therapeutic agent for cancer treatment. However, the mechanism of BV-induced apoptosis death should be clarified at the molecular level.


Assuntos
Antineoplásicos , Venenos de Abelha , Neoplasias de Mama Triplo Negativas , Humanos , Abelhas , Animais , Venenos de Abelha/farmacologia , Venenos de Abelha/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose , Linhagem Celular Tumoral
16.
Toxicon ; 222: 106993, 2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36528210

RESUMO

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.


Assuntos
Venenos de Abelha , Glioblastoma , Canais de Cátion TRPM , Humanos , Cisplatino/farmacologia , Cisplatino/metabolismo , Glioblastoma/tratamento farmacológico , Estresse Oxidativo , Meliteno/farmacologia , Canais de Cátion TRPM/metabolismo , Venenos de Abelha/farmacologia , Peróxido de Hidrogênio/farmacologia , Peróxido de Hidrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Apoptose , Oxidantes/farmacologia , Cálcio/metabolismo
17.
Toxins (Basel) ; 14(12)2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36548715

RESUMO

The venom of honeybees is composed of numerous peptides and proteins and has been used for decades as an anti-inflammatory and anti-cancer agent in traditional medicine. However, the bioactivity of specific biomolecular components has been evaluated for the predominant constituent, melittin. So far, only a few melittin-like peptides from solitary bee species have been investigated, and the molecular mechanisms of bee venoms as therapeutic agents remain largely unknown. Here, the preclinical pharmacological activities of known and proteo-transcriptomically discovered new melittin variants from the honeybee and more ancestral variants from phylogenetically older solitary bees were explored in the context of cancer and inflammation. We studied the effects of melittin peptides on cytotoxicity, second messenger release, and inflammatory markers using primary human cells, non-cancer, and cancerous cell lines. Melittin and some of its variants showed cytotoxic effects, induced Ca2+ signaling and inhibited cAMP production, and prevented LPS-induced NO synthesis but did not affect the IP3 signaling and pro-inflammatory activation of endothelial cells. Compared to the originally-described melittin, some phylogenetically more ancestral variants from solitary bees offer potential therapeutic modalities in modulating the in vitro inflammatory processes, and hindering cancer cell viability/proliferation, including aggressive breast cancers, and are worth further investigation.


Assuntos
Anti-Inflamatórios , Antineoplásicos , Venenos de Abelha , Abelhas , Meliteno , Animais , Humanos , Venenos de Abelha/farmacologia , Venenos de Abelha/química , Células Endoteliais , Meliteno/química , Meliteno/isolamento & purificação , Meliteno/farmacologia , Anti-Inflamatórios/química , Anti-Inflamatórios/isolamento & purificação , Anti-Inflamatórios/farmacologia , Antineoplásicos/química , Antineoplásicos/isolamento & purificação , Antineoplásicos/farmacologia , Linhagem Celular Tumoral
18.
PLoS One ; 17(8): e0272776, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35947632

RESUMO

Hepatocellular carcinoma (HCC) is one of the world's most risky diseases due to the lack of clear and cost-effective therapeutic targets. Currently, the toxicity of conventional chemotherapeutic medications and the development of multidrug resistance is driving research into targeted therapies. The nano-biomedical field's potential for developing an effective therapeutic nano-sized drug delivery system is viewed as a significant pharmaceutical trend for the encapsulation and release of numerous anticancer therapies. In this regard, current research is centered on the creation of biodegradable chitosan nanoparticles (CSNPs) for the selective and sustained release of bee venom into liver cancer cells. Furthermore, surface modification with polyethylene glycol (PEG) and GE11 peptide-conjugated bee venom-CSNPs allows for the targeting of EGFR-overexpressed liver cancer cells. A series of in vitro and in vivo cellular analyses were used to investigate the antitumor effects and mechanisms of targeted bee venom-CSNPs. Targeted bee venom-CSNPs, in particular, were found to have higher cytotoxicity against HepG2 cells than SMMC-7721 cells, as well as stronger cellular uptake and a substantial reduction in cell migration, leading to improved cancer suppression. It also promotes cancer cell death in EGFR overexpressed HepG2 cells by boosting reactive oxygen species, activating mitochondria-dependent pathways, inhibiting EGFR-stimulated MEK/ERK pathway, and elevating p38-MAPK in comparison to native bee venom. In hepatocellular carcinoma (HCC)-induced mice, it has anti-cancer properties against tumor tissue. It also improved liver function and architecture without causing any noticeable toxic side effects, as well as inhibiting tumor growth by activating the apoptotic pathway. The design of this cancer-targeted nanoparticle establishes GE11-bee venom-CSNPs as a potential chemotherapeutic treatment for EGFR over-expressed malignancies. Finally, our work elucidates the molecular mechanism underlying the anticancer selectivity of targeted bee venom-CSNPs and outlines therapeutic strategies to target liver cancer.


Assuntos
Venenos de Abelha , Carcinoma Hepatocelular , Quitosana , Neoplasias Hepáticas , Nanopartículas , Animais , Venenos de Abelha/farmacologia , Venenos de Abelha/uso terapêutico , Carcinoma Hepatocelular/patologia , Quitosana/uso terapêutico , Receptores ErbB/metabolismo , Neoplasias Hepáticas/patologia , Sistema de Sinalização das MAP Quinases , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Peptídeos/metabolismo
19.
Iran J Med Sci ; 47(4): 300-313, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35919080

RESUMO

Venomous arthropods such as scorpions and bees form one of the important groups with an essential role in medical entomology. Their venom possesses a mixture of diverse compounds, such as peptides, some of which have toxic effects, and enzymatic peptide Phospholipase A2 (PLA2) with a pharmacological potential in the treatment of a wide range of diseases. Bee and scorpion venom PLA2 group III has been used in immunotherapy, the treatment of neurodegenerative and inflammatory diseases. They were assessed for antinociceptive, wound healing, anti-cancer, anti-viral, anti-bacterial, anti-parasitic, and anti-angiogenesis effects. PLA2 has been identified in different species of scorpions and bees. The anti-leishmania, anti-bacterial, anti-viral, and anti-malarial activities of scorpion PLA2 still need further investigation. Many pieces of research have been stopped in the laboratory stage, and several studies need vast investigation in the clinical phase to show the pharmacological potential of PLA2. In this review, the medical significance of PLA2 from the venom of two arthropods, namely bees and scorpions, is discussed.


Assuntos
Venenos de Abelha , Venenos de Escorpião , Animais , Venenos de Abelha/química , Venenos de Abelha/farmacologia , Venenos de Abelha/uso terapêutico , Abelhas , Peptídeos , Fosfolipases A2/química , Fosfolipases A2/farmacologia , Fosfolipases A2/uso terapêutico , Venenos de Escorpião/farmacologia , Venenos de Escorpião/uso terapêutico , Escorpiões
20.
Cells ; 11(12)2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35741057

RESUMO

Alcohols are a part of cellular metabolism, but their physiological roles are not well understood. We investigated the effects of short-chain alcohols on Daphnia pulex and model membranes mimicking the lipid composition of eukaryotic inner mitochondrial membranes. We also studied the synergistic effects of alcohols with the bee venom membrane-active peptide, melittin, which is structurally similar to endogenous membrane-active peptides. The alcohols, from ethanol to octanol, gradually decreased the heart rate and the mitochondrial ATP synthesis of daphnia; in contrast, in combination with melittin, which exerted no sizeable effect, they gradually increased both the heart rate and the ATP synthesis. Lipid packing and the order parameter of oriented films, monitored by EPR spectroscopy of the spin-labeled probe 5-doxylstrearic acid, revealed gradual alcohol-assisted bilayer to non-bilayer transitions in the presence of melittin; further, while the alcohols decreased, in combination with melittin they increased the order parameter of the film, which is attributed to the alcohol-facilitated association of melittin with the membrane. A 1H-NMR spectroscopy of the liposomes confirmed the enhanced induction of a non-bilayer lipid phase that formed around the melittin, without the permeabilization of the liposomal membrane. Our data suggest that short-chain alcohols, in combination with endogenous peptides, regulate protein functions via modulating the lipid polymorphism of membranes.


Assuntos
Venenos de Abelha , Meliteno , Trifosfato de Adenosina , Álcoois/farmacologia , Venenos de Abelha/farmacologia , Lipídeos , Lipossomos , Meliteno/química , Meliteno/metabolismo , Meliteno/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA