Seyed Sadegh Eslami, Davod Jafari, Abbas Ghotaslou, Moein Amoupour, Amir Asri Kojabad, Rasool Jafari, Navid Mousazadeh, Parastoo Tarighi, Majid Sadeghizadeh
Purpose: Chemotherapy drugs used to treat lung cancer are associated with drug resistance and severe side effects. There have been rising demands for new therapeutic candidates and novel approaches, including combination therapy. Here, we aimed to investigate the combinatorial effect of a dendrosomal formulation of curcumin (DNC) and daunorubicin (DNR) on the A549 lung cancer cell line.
Methods: We performed cytotoxicity, apoptosis, cell migration, colony-formation capacity, and gene expression analysis to interpret the mechanism of action for a combination of DNC and DNR on A549 cells.
Results: Our results revealed that the combination of DNC and DNR could synergistically inhibit the A549 cells' growth. This synergistic cytotoxicity was further approved by flow cytometry, migration assessment, colony-forming capacity and gene expression analysis. DNR combination with DNC resulted in increased apoptosis to necrosis ratio compared to DNR alone. In addition, the migration and colony-forming capacity were at the minimal range when DNC was combined with DNR. Combined treatment decreased the expression level of MDR-1, hTERT and Bcl-2 genes significantly. In addition, the ratio of Bax/Bcl2 gene expression significantly increased. Our analysis by free curcumin, dendrosomes and DNC also showed that dendrosomes do not have any significant cytotoxic effect on the A549 cells, suggesting that this carrier has a high potential for enhancing the curcumin's biological effects.
Conclusion: Our observations suggest that the DNC formulation of curcumin synergistically enhances the antineoplastic effect of DNR on the A549 cell line through the modulation of apoptosis/necrosis ratio, as well as Bax/Bcl2 ratio, MDR-1 and hTERT gene expression.
{"title":"Combined Treatment of Dendrosomal-Curcumin and Daunorubicin Synergistically Inhibit Cell Proliferation, Migration and Induce Apoptosis in A549 Lung Cancer Cells.","authors":"Seyed Sadegh Eslami, Davod Jafari, Abbas Ghotaslou, Moein Amoupour, Amir Asri Kojabad, Rasool Jafari, Navid Mousazadeh, Parastoo Tarighi, Majid Sadeghizadeh","doi":"10.34172/apb.2023.050","DOIUrl":"https://doi.org/10.34172/apb.2023.050","url":null,"abstract":"<p><strong>Purpose: </strong>Chemotherapy drugs used to treat lung cancer are associated with drug resistance and severe side effects. There have been rising demands for new therapeutic candidates and novel approaches, including combination therapy. Here, we aimed to investigate the combinatorial effect of a dendrosomal formulation of curcumin (DNC) and daunorubicin (DNR) on the A549 lung cancer cell line.</p><p><strong>Methods: </strong>We performed cytotoxicity, apoptosis, cell migration, colony-formation capacity, and gene expression analysis to interpret the mechanism of action for a combination of DNC and DNR on A549 cells.</p><p><strong>Results: </strong>Our results revealed that the combination of DNC and DNR could synergistically inhibit the A549 cells' growth. This synergistic cytotoxicity was further approved by flow cytometry, migration assessment, colony-forming capacity and gene expression analysis. DNR combination with DNC resulted in increased apoptosis to necrosis ratio compared to DNR alone. In addition, the migration and colony-forming capacity were at the minimal range when DNC was combined with DNR. Combined treatment decreased the expression level of <i>MDR-1, hTERT</i> and <i>Bcl-2</i> genes significantly. In addition, the ratio of <i>Bax/Bcl2</i> gene expression significantly increased. Our analysis by free curcumin, dendrosomes and DNC also showed that dendrosomes do not have any significant cytotoxic effect on the A549 cells, suggesting that this carrier has a high potential for enhancing the curcumin's biological effects.</p><p><strong>Conclusion: </strong>Our observations suggest that the DNC formulation of curcumin synergistically enhances the antineoplastic effect of DNR on the A549 cell line through the modulation of apoptosis/necrosis ratio, as well as <i>Bax/Bcl2</i> ratio, <i>MDR-1</i> and <i>hTERT</i> gene expression.</p>","PeriodicalId":7256,"journal":{"name":"Advanced pharmaceutical bulletin","volume":"13 3","pages":"539-550"},"PeriodicalIF":3.6,"publicationDate":"2023-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10460814/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10176423","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Purpose: The phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/ mTOR) pathway is a complex intracellular metabolic pathway that leads to cell growth and tumor proliferation and plays a key role in drug resistance in breast cancer. Therefore, the anti-cancer effects of oleanolic acid (OA), maslinic acid (MA), and their combination were investigated to improve the performance of the treatment strategy.
Methods: We investigated the effect of OA and MA on cell viability using the WST-1 method. The synergistic effect of the combination was analyzed by isobologram analysis. In addition, the effects of the two compounds, individually and in combination, on apoptosis, autophagy, and the cell cycle were investigated in MCF7 cells. In addition, changes in the expression of PI3K/AKT/mTOR genes involved in apoptosis, cell cycle and metabolism were determined by quantitative RT-PCR.
Results: MA, OA, and a combination of both caused G0/G1 arrest. Apoptosis also increased in all treated groups. The autophagosomal LC3-II formation was induced 1.74-fold in the MA-treated group and 3.25-fold in the MA-OA-treated group. The combination treatment resulted in increased expression of genes such as GSK3B, PTEN, CDKN1B and FOXO3 and decreased expression of IGF1, PRKCB and AKT3 genes.
Conclusion: The results showed that the combination of these two substances showed the highest synergistic effect at the lowest dose and using MA-OA caused cancer cells to undergo apoptosis. The use of combination drugs may reduce the resistance of cancer cells to treatment.
{"title":"Enhanced Anti-cancer Potency Using a Combination of Oleanolic Acid and Maslinic Acid to Control Treatment Resistance in Breast Cancer.","authors":"Cigir Biray Avcı, Fatma Sogutlu, Neslihan Pinar Ozates, Behrouz Shademan, Cumhur Gunduz","doi":"10.34172/apb.2023.057","DOIUrl":"https://doi.org/10.34172/apb.2023.057","url":null,"abstract":"<p><strong>Purpose: </strong>The phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/ mTOR) pathway is a complex intracellular metabolic pathway that leads to cell growth and tumor proliferation and plays a key role in drug resistance in breast cancer. Therefore, the anti-cancer effects of oleanolic acid (OA), maslinic acid (MA), and their combination were investigated to improve the performance of the treatment strategy.</p><p><strong>Methods: </strong>We investigated the effect of OA and MA on cell viability using the WST-1 method. The synergistic effect of the combination was analyzed by isobologram analysis. In addition, the effects of the two compounds, individually and in combination, on apoptosis, autophagy, and the cell cycle were investigated in MCF7 cells. In addition, changes in the expression of PI3K/AKT/mTOR genes involved in apoptosis, cell cycle and metabolism were determined by quantitative RT-PCR.</p><p><strong>Results: </strong>MA, OA, and a combination of both caused G0/G1 arrest. Apoptosis also increased in all treated groups. The autophagosomal LC3-II formation was induced 1.74-fold in the MA-treated group and 3.25-fold in the MA-OA-treated group. The combination treatment resulted in increased expression of genes such as GSK3B, PTEN, CDKN1B and FOXO3 and decreased expression of IGF1, PRKCB and AKT3 genes.</p><p><strong>Conclusion: </strong>The results showed that the combination of these two substances showed the highest synergistic effect at the lowest dose and using MA-OA caused cancer cells to undergo apoptosis. The use of combination drugs may reduce the resistance of cancer cells to treatment.</p>","PeriodicalId":7256,"journal":{"name":"Advanced pharmaceutical bulletin","volume":"13 3","pages":"611-620"},"PeriodicalIF":3.6,"publicationDate":"2023-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10460813/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10112022","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neurodegenerative diseases are comprise a prominent class of neurological diseases. Generally, neurodegenerative diseases cannot be cured, and the available treatments can only regulate the symptoms or delay the disease progression. Among the several factors which could clarify the possible pathogenesis of neurodegenerative diseases, next to aging as the main risk, the dietary related diseases are the most important. Vegetable oils, which are composed of triacyclglycerols as the main components and several other components in a trace amount, are the main part of our diet. This review aims to study the effect of refined or unrefined vegetable oil consumption as a preventive or aiding strategy to slow or halt the progression of neurodegenerative diseases. In the refining process, owing to the chemical materials or severe temperatures of the refining process, removal of the desirable minor components is sometimes unavoidable and thus a worrisome issue affecting physical and neurological health.
{"title":"Effect of Refined Edible Oils on Neurodegenerative Disorders.","authors":"Fataneh Hashempour-Baltork, Parastou Farshi, Adel Mirza Alizadeh, Sevda Eskandarzadeh, Solmaz Abedinzadeh, Sodeif Azadmard-Damirchi, Mohammadali Torbati","doi":"10.34172/apb.2023.060","DOIUrl":"https://doi.org/10.34172/apb.2023.060","url":null,"abstract":"<p><p>Neurodegenerative diseases are comprise a prominent class of neurological diseases. Generally, neurodegenerative diseases cannot be cured, and the available treatments can only regulate the symptoms or delay the disease progression. Among the several factors which could clarify the possible pathogenesis of neurodegenerative diseases, next to aging as the main risk, the dietary related diseases are the most important. Vegetable oils, which are composed of triacyclglycerols as the main components and several other components in a trace amount, are the main part of our diet. This review aims to study the effect of refined or unrefined vegetable oil consumption as a preventive or aiding strategy to slow or halt the progression of neurodegenerative diseases. In the refining process, owing to the chemical materials or severe temperatures of the refining process, removal of the desirable minor components is sometimes unavoidable and thus a worrisome issue affecting physical and neurological health.</p>","PeriodicalId":7256,"journal":{"name":"Advanced pharmaceutical bulletin","volume":"13 3","pages":"461-468"},"PeriodicalIF":3.6,"publicationDate":"2023-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10460797/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10121089","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bhagyalakshmi Nair, Anisha Kuriakose, Bilha Baby, Lekshmi R Nath
Despite the significant improvement in the treatment modalities, cancer is one of the fastest-growing chronic disease conditions all over the world. Genetic and Epigenetic alterations in the normal physiology of the cell are the key factor for tumor development. These changes can trigger the production of abnormal protein expressions through stimulation of different signaling pathways and can deeply affect normal cell growth and proliferation. Any altered protein expression, genetic variation, micro-RNA or post-translational protein modifications that indicate tumorigenesis can act as an early signal termed as biomarker. Cancer, being a multistep process with accumulating genetic and epigenetic alterations, could be detected early with suitable biomarkers. There are several proteins such as AFP, CA-125, PSA, troponin, CEA, osteopontin, CA 19-9 that act as biomarkers which help in early detection, prognosis, and monitoring of disease progression, a hunt for newer biomarkers with higher specificity and sensitivity is still ongoing. Tumor-specific growth factor (TSGF) is one such budding and prevailing tumor biomarker used for the early-stage detection of several types of carcinomas. TSGF is a gene that helps in tumor angiogenesis and gets released during the preliminary stages from cancer cells that ensure the vascular proliferation of the same. In this review, the clinical investigations of TSGF in different kinds of malignancy is discussed in detail and suggests the possibility of using TSGF as a biomarker in early diagnosis of cancer.
{"title":"Tumor-Specific Growth Factor (TSGF): A Futuristic Tumor Biomarker in Early Diagnosis of Cancer.","authors":"Bhagyalakshmi Nair, Anisha Kuriakose, Bilha Baby, Lekshmi R Nath","doi":"10.34172/apb.2023.051","DOIUrl":"https://doi.org/10.34172/apb.2023.051","url":null,"abstract":"<p><p>Despite the significant improvement in the treatment modalities, cancer is one of the fastest-growing chronic disease conditions all over the world. Genetic and Epigenetic alterations in the normal physiology of the cell are the key factor for tumor development. These changes can trigger the production of abnormal protein expressions through stimulation of different signaling pathways and can deeply affect normal cell growth and proliferation. Any altered protein expression, genetic variation, micro-RNA or post-translational protein modifications that indicate tumorigenesis can act as an early signal termed as biomarker. Cancer, being a multistep process with accumulating genetic and epigenetic alterations, could be detected early with suitable biomarkers. There are several proteins such as AFP, CA-125, PSA, troponin, CEA, osteopontin, CA 19-9 that act as biomarkers which help in early detection, prognosis, and monitoring of disease progression, a hunt for newer biomarkers with higher specificity and sensitivity is still ongoing. Tumor-specific growth factor (TSGF) is one such budding and prevailing tumor biomarker used for the early-stage detection of several types of carcinomas. TSGF is a gene that helps in tumor angiogenesis and gets released during the preliminary stages from cancer cells that ensure the vascular proliferation of the same. In this review, the clinical investigations of TSGF in different kinds of malignancy is discussed in detail and suggests the possibility of using TSGF as a biomarker in early diagnosis of cancer.</p>","PeriodicalId":7256,"journal":{"name":"Advanced pharmaceutical bulletin","volume":"13 3","pages":"483-488"},"PeriodicalIF":3.6,"publicationDate":"2023-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10460812/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10123436","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Nadia Bolandi, Mohammad Hassan Khadem Ansari, Yousef Rasmi, Behzad Baradaran
Purpose: Despite the high prevalence of gastric cancer (GC), drug resistance is a major problem for effective chemotherapy. B7-H7 is a novel member of the B7 superfamily and is expressed in most common cancers. However, the role of B7-H7 on the aggressiveness of GC and chemosensitivity has remained unknown. Therefore, this study was designed to assess the effect of B7-H7 suppression using small interference RNA (siRNA) in combination with docetaxel on GC cells.
Methods: MTT test was applied to determine the IC50 of docetaxel and the combined effect of B7-H7 siRNA and docetaxel on the viability of the MKN-45 cells. To determine B7-H7, BCL-2, BAX, and caspase-3-8-9 genes expression, qRT-PCR was performed. Furthermore, flow cytometry was applied to evaluate apoptosis and the cell cycle status. Finally, to evaluate the effect of this combination therapy on migratory capacity and colony-forming ability, wound healing assay and colony formation test were employed, respectively.
Results: B7-H7 suppression increased the chemo-sensitivity of MKN-45 cells to docetaxel. The expression of B7-H7 mRNA was reduced after using B7-H7 siRNA and docetaxel in MKN-45 GC cells. Also, B7-H7 suppression alongside docetaxel reduced cell migration and colony formation rate, arrested the cell cycle at the G2-M phase, and induced apoptosis by modulating the expression of apoptotic target genes.
Conclusion: B7-H7 plays a significant role in the chemo-sensitivity and pathogenesis of GC. Therefore, B7-H7 suppression, in combination with docetaxel, may be a promising therapeutic approach in treating GC.
{"title":"Cooperative Treatment of Gastric Cancer Using B7-H7 siRNA and Docetaxel; How Could They Modify Their Effectiveness?","authors":"Nadia Bolandi, Mohammad Hassan Khadem Ansari, Yousef Rasmi, Behzad Baradaran","doi":"10.34172/apb.2023.055","DOIUrl":"https://doi.org/10.34172/apb.2023.055","url":null,"abstract":"<p><strong>Purpose: </strong>Despite the high prevalence of gastric cancer (GC), drug resistance is a major problem for effective chemotherapy. B7-H7 is a novel member of the B7 superfamily and is expressed in most common cancers. However, the role of B7-H7 on the aggressiveness of GC and chemosensitivity has remained unknown. Therefore, this study was designed to assess the effect of B7-H7 suppression using small interference RNA (siRNA) in combination with docetaxel on GC cells.</p><p><strong>Methods: </strong>MTT test was applied to determine the IC50 of docetaxel and the combined effect of B7-H7 siRNA and docetaxel on the viability of the MKN-45 cells. To determine B7-H7, BCL-2, BAX, and caspase-3-8-9 genes expression, qRT-PCR was performed. Furthermore, flow cytometry was applied to evaluate apoptosis and the cell cycle status. Finally, to evaluate the effect of this combination therapy on migratory capacity and colony-forming ability, wound healing assay and colony formation test were employed, respectively.</p><p><strong>Results: </strong>B7-H7 suppression increased the chemo-sensitivity of MKN-45 cells to docetaxel. The expression of B7-H7 mRNA was reduced after using B7-H7 siRNA and docetaxel in MKN-45 GC cells. Also, B7-H7 suppression alongside docetaxel reduced cell migration and colony formation rate, arrested the cell cycle at the G2-M phase, and induced apoptosis by modulating the expression of apoptotic target genes.</p><p><strong>Conclusion: </strong>B7-H7 plays a significant role in the chemo-sensitivity and pathogenesis of GC. Therefore, B7-H7 suppression, in combination with docetaxel, may be a promising therapeutic approach in treating GC.</p>","PeriodicalId":7256,"journal":{"name":"Advanced pharmaceutical bulletin","volume":"13 3","pages":"573-582"},"PeriodicalIF":3.6,"publicationDate":"2023-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10460818/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10176429","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Purpose: Mesoporous silica nanoparticles (MSNs) have drawn substantial interest as drug nanocarriers for breast cancer therapy. Nevertheless, because of the hydrophilic surfaces, the loading of well-known hydrophobic polyphenol anticancer agent curcumin (Curc) into MSNs is usually very low. Methods: For this purpose, Curc molecules were loaded into amine-functionalized MSNs (MSNs-NH2 -Curc) and characterized using thermal gravimetric analysis (TGA), Fourier-transform infrared (FTIR), field emission scanning electron microscope (FE-SEM), transmission electron microscope (TEM), Brunauer-Emmett-Teller (BET). MTT assay and confocal microscopy, respectively, were used to determine the cytotoxicity and cellular uptake of the MSNs-NH2 - Curc in the MCF-7 breast cancer cells. Besides, the expression levels of apoptotic genes were evaluated via quantitative polymerase chain reaction (qPCR) and western blot. Results: It was revealed that MSNs-NH2 possessed high values of drug loading efficiency and exhibited slow and sustained drug release compared to bare MSNs. According to the MTT findings, while the MSNs-NH2 -Curc were nontoxic to the human non-tumorigenic MCF-10A cells at low concentrations, it could considerably decrease the viability of MCF-7 breast cancer cells compared to the free Curc in all concentrations after 24, 48 and 72 hours exposure times. A cellular uptake study using confocal fluorescence microscopy confirmed the higher cytotoxicity of MSNs-NH2 -Curc in MCF-7 cells. Further, it was found that the MSNs-NH2 -Curc could drastically affect the mRNA and protein levels of Bax, Bcl-2, caspase 3, caspase 9, and hTERT relative to the free Curc treatment. Conclusion: Taken together, these preliminary results suggest the amine-functionalized MSNs-based drug delivery platform as a promising alternative approach for Curc loading and safe breast cancer treatment.
{"title":"<i>In Vitro</i> Efficacy of Curcumin-Loaded Amine-Functionalized Mesoporous Silica Nanoparticles against MCF-7 Breast Cancer Cells.","authors":"Zahra Mohebian, Mirzaagha Babazadeh, Nosratollah Zarghami","doi":"10.34172/apb.2023.035","DOIUrl":"https://doi.org/10.34172/apb.2023.035","url":null,"abstract":"<p><p><b><i>Purpose:</i></b> Mesoporous silica nanoparticles (MSNs) have drawn substantial interest as drug nanocarriers for breast cancer therapy. Nevertheless, because of the hydrophilic surfaces, the loading of well-known hydrophobic polyphenol anticancer agent curcumin (Curc) into MSNs is usually very low. <b><i>Methods:</i></b> For this purpose, Curc molecules were loaded into amine-functionalized MSNs (MSNs-NH<sub>2</sub> -Curc) and characterized using thermal gravimetric analysis (TGA), Fourier-transform infrared (FTIR), field emission scanning electron microscope (FE-SEM), transmission electron microscope (TEM), Brunauer-Emmett-Teller (BET). MTT assay and confocal microscopy, respectively, were used to determine the cytotoxicity and cellular uptake of the MSNs-NH<sub>2</sub> - Curc in the MCF-7 breast cancer cells. Besides, the expression levels of apoptotic genes were evaluated via quantitative polymerase chain reaction (qPCR) and western blot. <b><i>Results:</i></b> It was revealed that MSNs-NH<sub>2</sub> possessed high values of drug loading efficiency and exhibited slow and sustained drug release compared to bare MSNs. According to the MTT findings, while the MSNs-NH<sub>2</sub> -Curc were nontoxic to the human non-tumorigenic MCF-10A cells at low concentrations, it could considerably decrease the viability of MCF-7 breast cancer cells compared to the free Curc in all concentrations after 24, 48 and 72 hours exposure times. A cellular uptake study using confocal fluorescence microscopy confirmed the higher cytotoxicity of MSNs-NH<sub>2</sub> -Curc in MCF-7 cells. Further, it was found that the MSNs-NH<sub>2</sub> -Curc could drastically affect the mRNA and protein levels of Bax, Bcl-2, caspase 3, caspase 9, and hTERT relative to the free Curc treatment. <b><i>Conclusion:</i></b> Taken together, these preliminary results suggest the amine-functionalized MSNs-based drug delivery platform as a promising alternative approach for Curc loading and safe breast cancer treatment.</p>","PeriodicalId":7256,"journal":{"name":"Advanced pharmaceutical bulletin","volume":"13 2","pages":"317-327"},"PeriodicalIF":3.6,"publicationDate":"2023-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10278223/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9764057","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Raziye Tajali, Akram Eidi, Hosein Ahmadi Tafti, Abdolreza Pazouki, Ali Mohammad Sharifi
Purpose: Insufficient angiogenesis is associated with serious diabetic complications. Recently, adipose-derived mesenchymal stem cells (ADScs) are known to be a promising tool causing therapeutic neovascularization. However, the overall therapeutic efficacy of these cells is impaired by diabetes. This study aims to investigate whether in vitro pharmacological priming with deferoxamine, a hypoxia mimetic agent, could restore the angiogenic potential of diabetic human ADSCs. Methods: Diabetic human ADSCs were treated with deferoxamine and compared to normal and nontreated diabetic ADSCs for the expression of hypoxia inducible factor 1-alpha (HIF-1α), vascular endothelial growth factor (VEGF), fibroblast growth factor-2 (FGF-2) and stromal cell-derived factor-1α (SDF-1α), at mRNA and protein levels, using qRT-PCR, western blotting and ELISA assay. Activities of matrix metalloproteinases (MMPs)-2 and -9 were measured using a gelatin zymography assay. Angiogenic potentials of conditioned media derived from normal, Deferoxamine treated, and non-treated ADSCs were determined by in vitro scratch assay and also three-dimensional tube formation assay. Results: It is demonstrated that deferoxamine (150 and 300 μM) stabilized HIF-1α in primed diabetic ADSCs. At the concentrations used, deferoxamine did not show any cytotoxic effects. In deferoxamine treated ADSCs, expression of VEGF, SDF-1α, FGF-2 and the activity of MMP-2 and MMP-9 were significantly increased compared to nontreated ADSCs. Moreover, deferoxamine increased the paracrine effects of diabetic ADSCs in promoting endothelial cell migration and tube formation. Conclusion: Deferoxamine might be an effective drug for pharmacological priming of diabetic ADSCs to enhance the expression of proangiogenic factors noted via HIF-1α accumulation. In addition, impaired angiogenic potential of conditioned medium derived from diabetic ADSCs was restored by deferoxamine.
{"title":"Restoring the Angiogenic Capacity of the Human Diabetic Adipose-derived mesenchymal stem cells Primed with Deferoxamine as a Hypoxia Mimetic Agent: Role of HIF-1α.","authors":"Raziye Tajali, Akram Eidi, Hosein Ahmadi Tafti, Abdolreza Pazouki, Ali Mohammad Sharifi","doi":"10.34172/apb.2023.021","DOIUrl":"https://doi.org/10.34172/apb.2023.021","url":null,"abstract":"<p><p><b><i>Purpose:</i></b> Insufficient angiogenesis is associated with serious diabetic complications. Recently, adipose-derived mesenchymal stem cells (ADScs) are known to be a promising tool causing therapeutic neovascularization. However, the overall therapeutic efficacy of these cells is impaired by diabetes. This study aims to investigate whether in vitro pharmacological priming with deferoxamine, a hypoxia mimetic agent, could restore the angiogenic potential of diabetic human ADSCs. <b><i>Methods:</i></b> Diabetic human ADSCs were treated with deferoxamine and compared to normal and nontreated diabetic ADSCs for the expression of hypoxia inducible factor 1-alpha (HIF-1α), vascular endothelial growth factor (VEGF), fibroblast growth factor-2 (FGF-2) and stromal cell-derived factor-1α (SDF-1α), at mRNA and protein levels, using qRT-PCR, western blotting and ELISA assay. Activities of matrix metalloproteinases (MMPs)-2 and -9 were measured using a gelatin zymography assay. Angiogenic potentials of conditioned media derived from normal, Deferoxamine treated, and non-treated ADSCs were determined by in vitro scratch assay and also three-dimensional tube formation assay. <b><i>Results:</i></b> It is demonstrated that deferoxamine (150 and 300 μM) stabilized HIF-1α in primed diabetic ADSCs. At the concentrations used, deferoxamine did not show any cytotoxic effects. In deferoxamine treated ADSCs, expression of VEGF, SDF-1α, FGF-2 and the activity of MMP-2 and MMP-9 were significantly increased compared to nontreated ADSCs. Moreover, deferoxamine increased the paracrine effects of diabetic ADSCs in promoting endothelial cell migration and tube formation. <b><i>Conclusion:</i></b> Deferoxamine might be an effective drug for pharmacological priming of diabetic ADSCs to enhance the expression of proangiogenic factors noted via HIF-1α accumulation. In addition, impaired angiogenic potential of conditioned medium derived from diabetic ADSCs was restored by deferoxamine.</p>","PeriodicalId":7256,"journal":{"name":"Advanced pharmaceutical bulletin","volume":"13 2","pages":"350-360"},"PeriodicalIF":3.6,"publicationDate":"2023-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10278222/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9764062","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Zeynab Ahmadi, Safar Farajnia, Davoud Farajzadeh, Naser Pouladi, Neda Pourvatan, Mohammad Karbalaeimahdi, Fahime Shayegh, Maryam Arya
Purpose: The human somatropin is a single-chain polypeptide with a pivotal role in various biological processes. Although Escherichia coli is considered as a preferred host for the production of human somatropin, the high expression of this protein in E. coli results in the accumulation of protein as inclusion bodies. Periplasmic expression using signal peptides could be used to overcome the formation of inclusion bodies; still, the efficiency of each of the signal peptides in periplasmic transportation is varied and often is protein specific. The present study aimed to use in silico analysis to identify an appropriate signal peptide for the periplasmic expression of human somatropin in E. coli. Methods: A library containing 90 prokaryotic and eukaryotic signal peptides were collected from the signal peptide database, and each signal's characteristics and efficiency in connection with the target protein were analyzed by different software. The prediction of the secretory pathway and the cleavage position was determined by the signalP5 server. Physicochemical properties, including molecular weight, instability index, gravity, and aliphatic index, were investigated by ProtParam software. Results: The results of the present study showed that among all the signal peptides studied, five signal peptides ynfB, sfaS, lolA, glnH, and malE displayed high scores for periplasmic expression of human somatropin in E. coli, respectively. Conclusion: In conclusion, the results indicated that in-silico analysis could be used for the identification of suitable signal peptides for the periplasmic expression of proteins. Further laboratory studies can evaluate the accuracy of the results of in silico analysis.
{"title":"Optimized Signal Peptide for Secretory Expression of Human Recombinant Somatropin in <i>E. coli</i>.","authors":"Zeynab Ahmadi, Safar Farajnia, Davoud Farajzadeh, Naser Pouladi, Neda Pourvatan, Mohammad Karbalaeimahdi, Fahime Shayegh, Maryam Arya","doi":"10.34172/apb.2023.037","DOIUrl":"https://doi.org/10.34172/apb.2023.037","url":null,"abstract":"<p><p><b><i>Purpose:</i></b> The human somatropin is a single-chain polypeptide with a pivotal role in various biological processes. Although <i>Escherichia coli</i> is considered as a preferred host for the production of human somatropin, the high expression of this protein in <i>E. coli</i> results in the accumulation of protein as inclusion bodies. Periplasmic expression using signal peptides could be used to overcome the formation of inclusion bodies; still, the efficiency of each of the signal peptides in periplasmic transportation is varied and often is protein specific. The present study aimed to use <i>in silico</i> analysis to identify an appropriate signal peptide for the periplasmic expression of human somatropin in <i>E. coli</i>. <b><i>Methods:</i></b> A library containing 90 prokaryotic and eukaryotic signal peptides were collected from the signal peptide database, and each signal's characteristics and efficiency in connection with the target protein were analyzed by different software. The prediction of the secretory pathway and the cleavage position was determined by the signalP5 server. Physicochemical properties, including molecular weight, instability index, gravity, and aliphatic index, were investigated by ProtParam software. <b><i>Results:</i></b> The results of the present study showed that among all the signal peptides studied, five signal peptides ynfB, sfaS, lolA, glnH, and malE displayed high scores for periplasmic expression of human somatropin in <i>E. coli</i>, respectively. <b><i>Conclusion:</i></b> In conclusion, the results indicated that in-silico analysis could be used for the identification of suitable signal peptides for the periplasmic expression of proteins. Further laboratory studies can evaluate the accuracy of the results of <i>in silico</i> analysis.</p>","PeriodicalId":7256,"journal":{"name":"Advanced pharmaceutical bulletin","volume":"13 2","pages":"339-349"},"PeriodicalIF":3.6,"publicationDate":"2023-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10278209/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9764059","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Purpose: In the present study, we investigated the magnetic solid lipid nanoparticles (mSLNs) for targeted delivery of doxorubicin (DOX) into breast cancer cells. Methods: The synthesis of iron oxide nanoparticles was carried out by co-precipitation of a ferrous and ferric aqueous solution with the addition of a base; moreover, during precipitation process, the magnetite nanoparticles should be coated with stearic acid (SA) and tripalmitin (TPG). An emulsification dispersion-ultrasonic method was employed to prepare DOX loaded mSLNs. Fourier transforms infrared spectroscopy, vibrating sample magnetometer, and photon correlation spectroscopy (PCS) were used to characterize the subsequently prepared nanoparticles. In addition, the antitumor efficacy of particles was evaluated on MCF-7 cancer cell lines. Results: The findings showed that entrapment efficiency values for solid lipid and magnetic SLNs were 87±4.5% and 53.7±3.5%, respectively. PCS investigations showed that particle size increased with magnetic loading in the prepared NPs. In vitro drug release of DOX-loaded SLN and DOX-loaded mSLN in phosphate buffer saline (pH=7.4) showed that the amount of drug released approached 60% and 80%, respectively after 96 h of incubation. The electrostatic interactions between magnetite and drug had little effect on the release characteristics of the drug. The higher toxicity of DOX as nanoparticles compared to free drug was inferred from in vitro cytotoxicity. Conclusion: DOX encapsulated magnetic SLNs can act as a suitable and promising candidate for controlled and targeted therapy for cancer.
{"title":"Preparation and Characterization of Magnetic Solid Lipid Nanoparticles as a Targeted Drug Delivery System for Doxorubicin.","authors":"Abbas Soltani, Parvaneh Pakravan","doi":"10.34172/apb.2023.033","DOIUrl":"https://doi.org/10.34172/apb.2023.033","url":null,"abstract":"<p><p><b><i>Purpose:</i></b> In the present study, we investigated the magnetic solid lipid nanoparticles (mSLNs) for targeted delivery of doxorubicin (DOX) into breast cancer cells. <b><i>Methods:</i></b> The synthesis of iron oxide nanoparticles was carried out by co-precipitation of a ferrous and ferric aqueous solution with the addition of a base; moreover, during precipitation process, the magnetite nanoparticles should be coated with stearic acid (SA) and tripalmitin (TPG). An emulsification dispersion-ultrasonic method was employed to prepare DOX loaded mSLNs. Fourier transforms infrared spectroscopy, vibrating sample magnetometer, and photon correlation spectroscopy (PCS) were used to characterize the subsequently prepared nanoparticles. In addition, the antitumor efficacy of particles was evaluated on MCF-7 cancer cell lines. <b><i>Results:</i></b> The findings showed that entrapment efficiency values for solid lipid and magnetic SLNs were 87±4.5% and 53.7±3.5%, respectively. PCS investigations showed that particle size increased with magnetic loading in the prepared NPs. In vitro drug release of DOX-loaded SLN and DOX-loaded mSLN in phosphate buffer saline (pH=7.4) showed that the amount of drug released approached 60% and 80%, respectively after 96 h of incubation. The electrostatic interactions between magnetite and drug had little effect on the release characteristics of the drug. The higher toxicity of DOX as nanoparticles compared to free drug was inferred from <i>in vitro</i> cytotoxicity. <b><i>Conclusion:</i></b> DOX encapsulated magnetic SLNs can act as a suitable and promising candidate for controlled and targeted therapy for cancer.</p>","PeriodicalId":7256,"journal":{"name":"Advanced pharmaceutical bulletin","volume":"13 2","pages":"301-308"},"PeriodicalIF":3.6,"publicationDate":"2023-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10278217/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9701027","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Purpose: MicroRNAs (miRNAs) can contribute to cancer initiation, development, and progression. In this study, the effect of miRNA-4800 restoration on the growth and migration inhibition of human breast cancer (BC) cells was investigated. Methods: For this purpose, transfection of miR-4800 was performed into MDA-MB-231 BC cells using jetPEI. Subsequently, the expression levels of miR-4800 and CXCR4, ROCK1, CD44, and vimentin genes were measured using quantitative real-time polymerase chain reaction (q-RT-PCR) and specific primers. Also, the proliferation inhibition and apoptosis induction of cancer cells were evaluated by MTT and flow cytometry (Annexin V-PI method) techniques, respectively. Additionally, cancer cell migration after miR-4800 transfection was assessed by wound-healing (scratch) assay. Results: The restoration of miR-4800 in MDA-MB-231 cells resulted in the decreased expression level of CXCR4 (P ˂ 0.01), ROCK1 (P ˂ 0.0001), CD44 (P ˂ 0.0001), and vimentin (P ˂ 0.0001) genes. Also, MTT results showed restoration of miR-4800 could significantly reduce cell viability rate (P ˂ 0.0001) compared with the control group. Cell migration remarkably inhibited (P ˂ 0.001) upon miR-4800 transfection in treated BC cells. Flow cytometry data demonstrated that miR-4800 replacement considerably induced apoptosis in cancer cells (P ˂ 0.001) compared with control cells. Conclusion: Taken together, it seems that miR-4800 can act as a tumor suppressor miRNA in BC and play an essential role in modulating apoptosis, migration, and metastasis in BC. Therefore, it may be suggested as a potential therapeutic target in treating BC by performing additional tests in the future.
{"title":"The Effect of miR-4800 Restoration on Proliferation and Migration of Human Breast Cancer Cells <i>In Vitro</i>.","authors":"Monireh Khordadmehr, Reyhaneh Matin, Behzad Baradaran, Elham Baghbani, Farinaz Jigari-Asl, Saeed Noorolyai","doi":"10.34172/apb.2023.041","DOIUrl":"https://doi.org/10.34172/apb.2023.041","url":null,"abstract":"<p><p><b><i>Purpose:</i></b> MicroRNAs (miRNAs) can contribute to cancer initiation, development, and progression. In this study, the effect of miRNA-4800 restoration on the growth and migration inhibition of human breast cancer (BC) cells was investigated. <b><i>Methods:</i></b> For this purpose, transfection of miR-4800 was performed into MDA-MB-231 BC cells using jetPEI. Subsequently, the expression levels of miR-4800 and CXCR4, ROCK1, CD44, and vimentin genes were measured using quantitative real-time polymerase chain reaction (q-RT-PCR) and specific primers. Also, the proliferation inhibition and apoptosis induction of cancer cells were evaluated by MTT and flow cytometry (Annexin V-PI method) techniques, respectively. Additionally, cancer cell migration after miR-4800 transfection was assessed by wound-healing (scratch) assay. <b><i>Results:</i></b> The restoration of miR-4800 in MDA-MB-231 cells resulted in the decreased expression level of CXCR4 (<i>P</i> ˂ 0.01), ROCK1 (<i>P</i> ˂ 0.0001), CD44 (<i>P</i> ˂ 0.0001), and vimentin (<i>P</i> ˂ 0.0001) genes. Also, MTT results showed restoration of miR-4800 could significantly reduce cell viability rate (<i>P</i> ˂ 0.0001) compared with the control group. Cell migration remarkably inhibited (<i>P</i> ˂ 0.001) upon miR-4800 transfection in treated BC cells. Flow cytometry data demonstrated that miR-4800 replacement considerably induced apoptosis in cancer cells (<i>P</i> ˂ 0.001) compared with control cells. <b><i>Conclusion:</i></b> Taken together, it seems that miR-4800 can act as a tumor suppressor miRNA in BC and play an essential role in modulating apoptosis, migration, and metastasis in BC. Therefore, it may be suggested as a potential therapeutic target in treating BC by performing additional tests in the future.</p>","PeriodicalId":7256,"journal":{"name":"Advanced pharmaceutical bulletin","volume":"13 2","pages":"378-384"},"PeriodicalIF":3.6,"publicationDate":"2023-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10278211/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10067569","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}