14 results on '"Shanehbandi, Dariush"'
Search Results
2. Apoptosis Induction by New Coumarin Derivatives in a Mice Model of Breast Cancer.
- Author
-
Abbasi, Mehran Mesgari, Khordadmehr, Monireh, Shanehbandi, Dariush, Asl, Farinaz Jigari, Mofrad, Reza Teimuri, Tahmasebi, Shabnam, Asar, Mohammad Shahab, Vaezi, Fateme Eskandari, and Panahi, Yousef
- Subjects
COUMARINS ,COUMARIN derivatives ,BREAST cancer ,ANIMAL disease models ,LABORATORY mice ,CANCER cell growth ,BREAST - Abstract
In the last decades, numerous studies have focused on the search for new agents to suppress the growth of cancer cells. In this study, we investigated the effect of two novel synthetic coumarin derivatives, namely 2-amino-4- (4-(2-hydroxyethoxy)-3-methoxyphenyl)-5-oxo-4H,5H-pyrano[3,2-c]coumarin-3-carbonitrile and 2-amino-4-(4-hydroxyphenyl)-5-oxo-4H,5H-pyrano[3,2-c]coumarin-3-carbonitrile, on the induction of apoptosis in breast cancer in a mouse model. Breast cancer was induced in BALB/c mice, which were randomly divided into six groups and then underwent the experiment. The groups and treatments included A1 : coumarin A with a low dose (10 µm), A2: coumarin A with a high dose (1 mM), B1: coumarin B with a low dose (10 µm), B2: coumarin B with a high dose (1 mM), D: doxorubicin, and C: cancer control/ treatment with normal saline. The samples underwent treatments for 5 weeks. Animals were euthanized, and tissue samples, including the lung, liver, and tumor mass, were collected for histopathological examination. In addition, quantitative real-time polymerase chain reaction (qRT-PCR) was performed to determine some apoptotic markers, such as BCL-2, caspase-9, COX-2, and c-Myc. The qRT- PCR presented that both coumarin compounds could significantly alter the expression levels of BCL-2, caspase-9, COX-2, and c-Myc. Consistent with these results, histopathological observations showed a significant reduction in pathological lesions and severity of malignancy of the tumor mass, as well as a decrease in microscopic metastases in the lung and liver. This suggests that the present new coumarin compounds may induce apoptosis in breast cancer cells by altering some apoptosis-related genes that may play a chemotherapeutic role in breast cancer therapy in the future. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
3. Restoring of miR-193a-5p Sensitizes Breast Cancer Cells to Paclitaxel through P53 Pathway.
- Author
-
Khordadmehr, Monireh, Shahbazi, Roya, Baradaran, Behzad, Sadreddini, Sanam, Shanehbandi, Dariush, and Hajiasgharzadeh, Khalil
- Subjects
CANCER cells ,PACLITAXEL ,BREAST cancer ,P53 antioncogene ,APOPTOSIS ,MICRORNA ,CELL survival ,DRUG resistance in cancer cells - Abstract
Purpose: Recent evidence presented the important role of microRNAs in health and disease particularly in human cancers. Among those, miR-193 family contributes as a tumor suppressor in different benign and malignant cancers like breast cancer (BC) via interaction with specific targets. On the other hand, it was stated that miR-193 is able to modulate some targets in chemoresistant cancer cells. Therefore, the aim of this study was to evaluate the potential function of miR-193a-5p and paclitaxel in the apoptosis induction by targeting P53 in BC cells. Methods: At first, miR-193a-5p mimics were transfected to MDA-MB-231 BC cell line which indicated the lower expression level of miR-193a-5p. Subsequently, the transfected cells were treated with paclitaxel. Then, cell viability, apoptosis, and migration were evaluated by MTT, flow cytometry and DAPI staining, and scratch-wound motility assays, respectively. Moreover, the expression levels of P53 was evaluated by qRT-PCR. Results: The expression level of miR-193a-5p was restored in MDA-MB-231 cells which profoundly inhibited the proliferation (P < 0.0001), induced apoptosis (P < 0.0001) and harnessed migration (P < 0.0001) in the BC cells and more effectiveness was observed in combination with paclitaxel. Interestingly, increased miR-193a-5p expression led to a reduction in P53 mRNA, offering that it can be a potential target of miR-193a. Conclusion: Taken together, it is concluded that the combination of miR-193a-5p restoration and paclitaxel could be potentially considered as an effective therapeutic strategy to get over chemoresistance during paclitaxel chemotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
4. Paclitaxel May Inhibit Epithelial-Mesenchymal Transition Properties of Triple-negative Breast Cancer Cell Line via Altering the Expression of EMT-promoting and --inhibiting MicroRNAs.
- Author
-
Haghnavaz, Navideh, Asghari, Faezeh, Shekari, Najibeh, Shanehbandi, Dariush, Javadian, Mahsa, Mohammadi, Ali, Baradaran, Behzad, and Kazemi, Tohid
- Subjects
ANTINEOPLASTIC agents ,BREAST tumors ,CELL culture ,CELL lines ,CYTOSKELETAL proteins ,DNA ,GENE expression ,PACLITAXEL ,POLYMERASE chain reaction ,RNA ,QUANTITATIVE research ,TREATMENT effectiveness ,MICRORNA ,MATRIX metalloproteinases ,PHARMACODYNAMICS ,EVALUATION - Abstract
Background: Abnormal expressions of microRNAs are related to various cancers such as breast cancer for which paclitaxel is widely used as a chemotherapeutic agent. We aimed to investigate the effect of paclitaxel treatment on the expression level of miR-199a-5p and miR-10b, involved in epithelial-mesenchymal transition (EMT) process in breast cancer cell lines. Methods: Human breast cancer cell lines BT-474, SKBR-3, MDA-MB-231, and MCF-7 were cultured and MTT assay was used to determine IC50 of paclitaxel. RNA was extracted, cDNA was synthesized, and the expression level of miRNAs and genes was quantitatively determined using real-time PCR. Results: After treatment with paclitaxel, the expression level of miR-199a-5p significantly decreased in MCF-7 and SKBR-3 cell lines, while it increased in MDAMB-231 and BT-474. The expression level of miR-10b was also significantly reduced in MCF-7, MDA-MB-231, and SKBR-3 and increased in BT-474 cell lines following treatment with paclitaxel. Our results further indicated that paclitaxel reduced the expression level of vimentin and MMP-9 in MDA-MB-231 cell line. Conclusion: Our findings revealed the increased expression of EMT-inhibitor miR-199a-5p and the decreased expression of metastamir miR-10b after treatment of MDA-MB-231 metastatic breast cancer cell line. Reduced expressions of vimentin and MMP-9 were also observed, corroborating the inhibition of metastasis markers in this type of breast cancer. The therapeutic effect of paclitaxel may in part be due to the change in the balance of EMT-promoting and EMT-inhibiting miRNAs. [ABSTRACT FROM AUTHOR]
- Published
- 2020
5. Targeted Co-Delivery of Docetaxel and cMET siRNA for Treatment of Mucin1 Overexpressing Breast Cancer Cells.
- Author
-
Majidi Zolbanin, Naime, Jafari, Reza, Majidi, Jafar, Atyabi, Fatemeh, Yousefi, Mehdi, Jadidi-Niaragh, Farhad, Aghebati-Maleki, Leili, Shanehbandi, Dariush, Soltani Zangbar, Mohammad-Sadegh, and Nayebi, Alireza Mohajjel
- Subjects
BREAST cancer ,CANCER cells ,ELECTROPHORESIS ,GENE expression ,DRUG delivery systems ,NANOPARTICLES ,DOCETAXEL - Abstract
Purpose: Targeted treatment of breast cancer through combination of chemotherapeutic agents and siRNA had been drawing much attention in recent researches. This study was carried out to evaluate mucin1 aptamer-conjugated chitosan nanoparticles containing docetaxel and cMET siRNA on SKBR3 cells. Methods: Nano-drugs were characterized by transmission electron microscope, Zetasizer and loading efficiency calculation. siRNA entrapment onto nanoparticles, stability of siRNA-loaded nanoparticles and conjugation of mucin1 aptamer to nanoparticles were evaluated via separate electrophoresis. Cellular uptake of the targeted nanoparticles was evaluated through GFP-plasmid expression in mucin1+ SKBR3 vs. mucin1-CHO cells. Protein expression, cell viability and gene expression were assessed by Western Blotting, MTT assay, and Quantitative Real Time-PCR, respectively. Results: Characterization of nano-drugs represented the ideal size (110.5± 3.9 nm), zeta potential (11.6± 0.8 mV), and loading efficiency of 90.7% and 88.3% for siRNA and docetaxel, respectively. Different gel electrophoresis affirmed the conjugation of aptamers to nanoparticles and entrapment of siRNA onto nanoparticles. Increased cellular uptake of aptamer-conjugated nanoparticles was confirmed by GFP expression. cMET gene silencing was confirmed by Western Blotting. The significant (p ≤0.0001) impact of combination targeted therapy vs. control on cell viability was shown. Results of Quantitative Real Time-PCR represented a remarkably decreased (p ≤0.0001) expression of the studied genes involving in tumorigenicity, metastasis, invasion, and angiogenesis (STAT3, IL8, MMP2, MMP9, and VEGF) by targeted combination treatment vs. control. Conclusion: The mucin1 aptamer-conjugated chitosan nanoparticles, containing docetaxel and cMET siRNA, is suggested for treatment of mucin1+ metastatic breast cancer cells. However, further studies should be conducted on animal models. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
6. HER2 positivity may confer resistance to therapy with paclitaxel in breast cancer cell lines.
- Author
-
Haghnavaz, Navideh, Asghari, Faezeh, Elieh Ali Komi, Daniel, Shanehbandi, Dariush, Baradaran, Behzad, and Kazemi, Tohid
- Subjects
CANCER cells ,CELL lines ,PACLITAXEL ,GENETICS of breast cancer ,MICRORNA - Abstract
Introduction: MicroRNAs (miRNAs) are short non-coding single-stranded RNAs. Involving in post-transcriptional gene silencing, miRNAs are thought to play important roles in many cancers such as breast cancer. Paclitaxel is used widely in the treatment of breast cancer. In this study, we investigated the effect of paclitaxel treatment on the expression levels of two oncomirs (oncomiRs), miR-21 and miR-203, in breast cancer cell lines.Materials and methods: MTT assay was performed to determine IC50 of paclitaxel for human breast cancer cell lines including MCF-7, MDA-MB-231, SKBR3 and BT-474. After RNA extraction and cDNA synthesis, the expression levels of miRNAs were then quantitatively evaluated using real-time PCR.Results: Our results showed that after treatment, the expression levels of both miR-21 and miR-203 were significantly increased in HER2-positive cell lines, BT-474 and SKBR3. HER2-negative cell lines, MCF-7 and MDA-MB-231, in contrast had significantly decreased expression of both assessed oncomiRs.Conclusion: Our results showed that the expression levels of oncomiRs were increased in HER-2 positive breast cancer cells and this finding is in line with previous studies. Our findings present a probable mechanism of resistance against paclitaxel chemotherapy in HER2-positive breast cancers. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
7. The Cytotoxic and Apoptotic Effects of Scrophularia Atropatana Extracts on Human Breast Cancer Cells.
- Author
-
Safarzadeh, Elham, Delazar, Abbas, Kazemi, Tohid, Orangi, Mona, Shanehbandi, Dariush, Esnaashari, Solmaz, Mohammadnejad, Leila, Sadigh-Eteghad, Saeed, mohammadi, Ali, Fakhr, Mehrdad Ghavifekr, and Baradaran, Behzad
- Subjects
FIGWORTS ,APOPTOSIS ,BREAST cancer ,CANCER cells ,PLANT extracts ,POLYMERASE chain reaction - Abstract
Purpose: Breast cancer is the most frequent malignancy diagnosed in women both in developed and developing countries. Natural products especially those from herbal origin have high potential in producing drug components with a source of novel structures. The present study was designed to explore the cytotoxic effects and the cell death mechanism of Scrophularia atropatana extracts. Methods: MTT assay was employed to evaluate the cytotoxic activity of the extracts of S. atropatana on the MCF-7 as well as non-malignant cells. Furthermore, induction of apoptosis was evaluated by TUNEL assay, cell death detection ELISA, DNA fragmentation test, western blotting and Real Time PCR. Results: In vitro exposures of the MCF-7 cells with different concentration of S. atropatana extract significantly inhibited their growth and viability and induced apoptosis in the MCF-7 cells. Cleavage PARP protein, decrease in the mRNA expression levels of bcl-2 and increase expression of Caspase-3 and Caspase-9 mRNA, highlights that the induction of apoptosis was the main mechanism of cell death. Moreover the expression study of Caspase-9 mRNA showed that, the extracts have induced apoptosis via intrinsic mitochondrial pathway. Conclusion: Our results demonstrated that dichloromethane extract of Scrophularia atropatana has an apoptotic effects and it can be developed as anticancer agents. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
8. MLN4924 and 2DG combined treatment enhances the efficiency of radiotherapy in breast cancer cells.
- Author
-
Oladghaffari, Maryam, Shabestani Monfared, Ali, Farajollahi, Alireza, Baradaran, Behzad, Mohammadi, Mohsen, Shanehbandi, Dariush, Asghari Jafar Abadi, Mohammad, and Pirayesh Islamian, Jalil
- Subjects
BREAST cancer treatment ,COMBINATION drug therapy ,RADIOTHERAPY ,TREATMENT effectiveness ,RADIATION-sensitizing agents ,CELL-mediated cytotoxicity ,APOPTOSIS - Abstract
Purpose:Two-deoxy-D-glucose (2DG) causes cytotoxicity in the cancer cells by disrupting the thiol metabolism, and MLN4924 inactivates the SCF E3 ligase and so causes the accumulation of its substrates which trigger apoptosis and hence might enhance the efficiency of radiotherapy and overcame on the radioresistance of the cancer cells. Materials and methods:SKBR3 and MCF-7 breast cancer cells were treated with 500 μM 2DG and/or MLN4924 (30, 100, 200 and 300 nM), and in combination in the presence and absence of 1, 1.5 and 2 Gy gamma irradiation. The effects of the treatments – 2DG, MLN4924, irradiation alone and combined – on MCF-7 and SKBR3 cell lines were evaluated by MTT assay, TUNEL assay, cell death detection, Q-PCR for caspase-3 and Bcl-2 expression analysis, and finally clonogenic survival assay. Results:The treatments enhanced the further radio cytotoxicity via inducing the apoptosis cell signaling gene, caspase-3. The 2DG and MLN4924 treatments could act as a radiosensitizer, especially on the SKBR3 cells, and further sensitized the cells with a sensitivity enhancement ratio (SER) of 1.41 and 1.27 in SKBR3 and MCF-7 cells, respectively. Conclusion:The combined chemo-radiotherapy might improve the breast cancer treatment outcome. [ABSTRACT FROM PUBLISHER]
- Published
- 2017
- Full Text
- View/download PDF
9. miR-34a and miR-200c Have an Additive Tumor-Suppressive Effect on Breast Cancer Cells and Patient Prognosis.
- Author
-
Mansoori, Behzad, Silvestris, Nicola, Mohammadi, Ali, Khaze, Vahid, Baghbani, Elham, Mokhtarzadeh, Ahad, Shanehbandi, Dariush, Derakhshani, Afshin, Duijf, Pascal H. G., and Baradaran, Behzad
- Subjects
BREAST cancer ,CANCER cells ,CANCER patients ,EPITHELIAL-mesenchymal transition ,CELL cycle - Abstract
Breast cancer is the most common women's malignancy in the world and, for subgroups of patients, treatment outcomes remain poor. Thus, more effective therapeutic strategies are urgently needed. MicroRNAs (miRNAs) have emerged as promising therapeutic tools and targets, as they play significant roles in regulating key cellular processes by suppressing gene expression. However, additive opportunities involving miRNAs have been underexplored. For example, both miR-34a and miR-200c individually suppress the development of different types of cancer, but the cellular effects of their combined actions remain unknown. Here, we show that miR-34a and miR-200c levels are reduced in breast tumors compared to adjacent normal tissues and that this additively predicts poor patient survival. In addition, in cell lines, miR-34a and miR-200c additively induce apoptosis and cell cycle arrest, while also inhibiting proliferation, invasion, migration, stemness and epithelial-to-mesenchymal transition (EMT). Mechanistically, both miRNA-34a and miR-200c directly target HIF1-α and subsequently downregulate VEGFR, MMP9 and CXCR4, although combined miRNA-34a and miR-200c delivery suppresses mouse xenograft tumor development as effectively as individual delivery. We establish a model, supported by in vitro and clinical data, which collectively suggest that the co-delivery of miR-34a and miR-200c represents a promising novel therapeutic strategy for breast cancer patients. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
10. Mir‐34a and mir‐200c have an additive tumor‐suppressive effect on breast cancer cells and patient prognosis
- Author
-
Behzad Baradaran, Pascal H.G. Duijf, Vahid Khaze, Nicola Silvestris, Behzad Mansoori, Dariush Shanehbandi, Ahad Mokhtarzadeh, Afshin Derakhshani, Ali Mohammadi, Elham Baghbani, Mansoori, Behzad, Silvestris, Nicola, Mohammadi, Ali, Khaze, Vahid, Baghbani, Elham, Mokhtarzadeh, Ahad, Shanehbandi, Dariush, Derakhshani, Afshin, Duijf, Pascal HG, and Baradaran, Behzad
- Subjects
0301 basic medicine ,cancer stemness ,Cell cycle checkpoint ,Apoptosis ,CXCR4 ,Metastasis ,miR-200c ,Mice ,0302 clinical medicine ,Breast cancer ,Cell Movement ,Medicine ,Genetics (clinical) ,apoptosis ,Middle Aged ,Prognosis ,Progression-Free Survival ,Gene Expression Regulation, Neoplastic ,Matrix Metalloproteinase 9 ,cell cycle arrest ,030220 oncology & carcinogenesis ,Heterografts ,Female ,miR-34a ,HIF1-α ,Receptors, CXCR4 ,Epithelial-Mesenchymal Transition ,lcsh:QH426-470 ,MiR‐200c ,Breast Neoplasms ,HIF1‐α ,Malignancy ,Article ,Cell cycle arrest ,03 medical and health sciences ,breast cancer ,Downregulation and upregulation ,Cell Line, Tumor ,microRNA ,Genetics ,Animals ,Humans ,metastasis ,Cancer stemness ,Cell Proliferation ,business.industry ,Cancer ,MiR‐34a ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,MicroRNAs ,lcsh:Genetics ,030104 developmental biology ,Cancer research ,business - Abstract
Breast cancer is the most common women’s malignancy in the world and, for subgroups ofpatients, treatment outcomes remain poor. Thus, more effective therapeutic strategies are urgently needed. MicroRNAs (miRNAs) have emerged as promising therapeutic tools and targets, as they play significant roles in regulating key cellular processes by suppressing gene expression. However, additive opportunities involving miRNAs have been underexplored. For example, both miR-34aand miR-200c individually suppress the development of different types of cancer, but the cellular effects of their combined actions remain unknown. Here, we show that miR-34a and miR-200c levels are reduced in breast tumors compared to adjacent normal tissues and that this additively predicts poor patient survival. In addition, in cell lines, miR-34a and miR-200c additively induce apoptosis and cell cycle arrest, while also inhibiting proliferation, invasion, migration, stemness andepithelial-to-mesenchymal transition (EMT). Mechanistically, both miRNA-34a and miR-200c directly target HIF1-α and subsequently downregulate VEGFR, MMP9 and CXCR4, although combined miRNA-34a and miR-200c delivery suppresses mouse xenograft tumor development as effectively as individual delivery. We establish a model, supported by in vitro and clinical data, which collectively suggest that the co-delivery of miR-34a and miR-200c represents a promising novel therapeuticstrategy for breast cancer patients. Breast cancer is the most common women’s malignancy in the world and, for subgroups of patients, treatment outcomes remain poor. Thus, more effective therapeutic strategies are urgently needed. MicroRNAs (miRNAs) have emerged as promising therapeutic tools and targets, as they play significant roles in regulating key cellular processes by suppressing gene expression. However, additive opportunities involving miRNAs have been underexplored. For example, both miR‐34a and miR‐200c individually suppress the development of different types of cancer, but the cellular effects of their combined actions remain unknown. Here, we show that miR‐34a and miR‐ 200c levels are reduced in breast tumors compared to adjacent normal tissues and that this additively predicts poor patient survival. In addition, in cell lines, miR‐34a and miR‐200c additively induce apoptosis and cell cycle arrest, while also inhibiting proliferation, invasion, migration, stemness and epithelial‐to‐mesenchymal transition (EMT). Mechanistically, both miRNA‐34a and miR‐ 200c directly target HIF1‐α and subsequently downregulate VEGFR, MMP9 and CXCR4, although combined miRNA‐34a and miR‐200c delivery suppresses mouse xenograft tumor development as effectively as individual delivery. We establish a model, supported by in vitro and clinical data, which collectively suggest that the co‐delivery of miR‐34a and miR‐200c represents a promising novel therapeutic strategy for breast cancer patients.
- Published
- 2021
- Full Text
- View/download PDF
11. Targeted delivery of doxorubicin by Thermo/pH-responsive magnetic nanoparticles in a rat model of breast cancer.
- Author
-
Pourradi, Nasibeh Mohammad Ali, Babaei, Hossein, Hamishehkar, Hamed, Baradaran, Behzad, Shokouhi-Gogani, Behrooz, Shanehbandi, Dariush, Ghorbani, Marjan, and Azarmi, Yadollah
- Subjects
- *
NANOMEDICINE , *MAGNETIC nanoparticles , *TARGETED drug delivery , *BREAST cancer , *DRUG delivery systems , *ANIMAL disease models , *POLYMERSOMES - Abstract
The novel folate conjugated Thermo/pH-responsive magnetic nanoparticles (folate-poly-MNPs) have been developed as a potential nanocarrier for improving site-specific drug delivery, tumor drug accumulation, and therapeutic effects while reducing the adverse effects of conventional drug delivery systems. To evaluate the anticancer efficacy of developed tumor-targeted drug delivery system, forty rat models of breast cancer received saline as control, DOX, DOX-poly-MNPs, and DOX-folate-poly-MNPs at a dose of 2 mg/kg/48 h. The DOX-folate-poly-MNPs showed a significant increase in protein expression of BAX and C-caspase-3 with concomitant downregulation of Bcl-2 expression and ki67 proliferation index compared to the DOX group. The synergistic antitumor efficacy of passive and active drug targeting led to enhanced drug uptake, increased tumor cell apoptosis, decreased tumor volume, and a prolonged survival rate in animals, suggesting that DOX-folate-poly-MNPs may prove to be a promising nanomedicine for the smart treatment of breast cancer in the future. [Display omitted] • Novel tumor-targeting DDSs improved DOX anticancer efficacy. • Tumor volume and ki67 proliferation index reduced in treated rats. • Body weight, BAX, C-caspase-3, and C-PARP1 expression increased in treated rats. • DOX-folate-poly-MNPs prolonged survival rates in breast cancer models. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
12. Overexpression of HMGA2 in breast cancer promotes cell proliferation, migration, invasion and stemness
- Author
-
Behzad Baradaran, Henrik J. Ditzel, Pascal H.G. Duijf, Khalil Hajiasgharzadeh, Elmira Roshani, Morten F. Gjerstorff, Dariush Shanehbandi, Solmaz Shirjang, Souzan Najafi, Behzad Mansoori, Ali Mohammadi, Ahad Mokhtarzadeh, Tohid Kazemi, Mansoori, Behzad, Duijf, Pascal HG, Mohammadi, Ali, Najafi, Souzan, Roshani, Elmira, Shanehbandi, Dariush, Hajiasgharzadeh, Khalil, Shirjang, Solmaz, Ditzel, Henrik J, Kazemi, Tohid, Mokhtarzadeh, Ahad, Gjerstorff, Morten F, and Baradaran, Behzad
- Subjects
0301 basic medicine ,Cell cycle checkpoint ,HMGA2 ,proliferation ,Clinical Biochemistry ,03 medical and health sciences ,stemness ,0302 clinical medicine ,Breast cancer ,breast cancer ,Cancer stem cell ,Drug Discovery ,medicine ,CD133 ,Pharmacology ,biology ,Cell growth ,apoptosis ,EMT ,Cancer ,Cell cycle ,medicine.disease ,030104 developmental biology ,Apoptosis ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Molecular Medicine ,cell cycle - Abstract
Despite improved therapeutic strategies for early-stage breast cancer, the most common cancer type in women, relapse remains common and the underlying mechanisms for this progression remain poorly understood. To gain more insight, we studied the DNA-binding protein HMGA2 in breast cancer development and stemness. We demonstrated that HMGA2 is overexpressed in breast cancer tissues at the mRNA and protein levels (P value
- Published
- 2020
- Full Text
- View/download PDF
13. PD-1/PD-L1 axis importance and tumor microenvironment immune cells.
- Author
-
Lotfinejad, Parisa, Kazemi, Tohid, Mokhtarzadeh, Ahad, Shanehbandi, Dariush, Jadidi Niaragh, Farhad, Safaei, Sahar, Asadi, Milad, and Baradaran, Behzad
- Subjects
- *
TRIPLE-negative breast cancer , *TUMOR microenvironment , *PROGRAMMED cell death 1 receptors , *BREAST cancer prognosis , *PROGRAMMED death-ligand 1 , *BREAST cancer - Abstract
Triple-negative breast cancer (TNBC) is heterogeneous cancer with poor prognosis among the other breast tumors. Rapid recurrence and increased progression rate could be reasons for the poor prognosis of this type of breast cancer. Recently, because of the lack of specific targets in multiple cancer treatment, immune checkpoint blockade therapies with targeting PD-1/PD-L1 axis have displayed significant advances and improved survival. Among different types of breast cancers, TNBC is considered more immunogenic with high T-cell and other immune cells infiltration compared to other breast cancer subtypes. This immunogenic characteristic of TNBC is a beneficial marker in the immunotherapy of these tumors. Clinical studies with a focus on immune checkpoint therapy have demonstrated promising results in TNBC treatment. In this review, we summarize clinical trials with the immunotherapy-based treatment of different cancers and also discuss the interaction between infiltrating immune cells and breast tumor microenvironment. In addition, we focus on the signaling pathway that controls PD-L1 expression and continues with CAR T-cell therapy and siRNA as novel strategies and potential tools in targeted therapy. Unlabelled Image • Negative Breast Cancer (TNBC) is more immunogenic in compare with other breast cancer types. • Immunotherapy is promising method in the management of patients with TNBC. • Whether the PD-L1 expression has a good prognostic effects on breast cancer outcome is controversial. • Immunogenicity of the tumor micro-environment is in favor of using targeted therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
14. Docosahexaenoic acid (DHA) inhibits pro-angiogenic effects of breast cancer cells via down-regulating cellular and exosomal expression of angiogenic genes and microRNAs.
- Author
-
Aslan, Cynthia, Maralbashi, Sepideh, Kahroba, Houman, Asadi, Milad, Soltani-Zangbar, Mohammad Sadegh, Javadian, Mahsa, Shanehbandi, Dariush, Baradaran, Behzad, Darabi, Masood, and Kazemi, Tohid
- Subjects
- *
EXOSOMES , *DOCOSAHEXAENOIC acid , *CANCER cells , *MICRORNA , *BREAST cancer , *FREE fatty acids - Abstract
Docosahexaenoic acid (DHA) as an omega 3 free fatty acid has been reported to exert anti-angiogenesis effects. However, our current understanding regarding the precise mechanisms of such effects is still limited. Exosomes secreted by cancer cells may act as angiogenesis promoters. The aim of the study was to determine altered expression levels of HIF-1α, TGF-β, VEGFR, Snail1, Snail2 and SOX2 and their regulating microRNAs in MDA-MB-231 and BT-474 cell lines after treatment with DHA in both normoxic and hypoxic conditions. Human breast cancer cell lines including MDA-MB-231 and BT-474 were treated for 24 h with 100 uM DHA under normoxic and hypoxic conditions. Exosomes were isolated from untreated and treated cells and characterized by transmission electron microscopy (TEM) and western blotting. RNAs from cells and isolated exosomes were extracted and cDNAs were synthesized. Expression levels of miRNAs and their pro-angiogenic target genes were analyzed using quantitative real-time PCR (qRT-PCR). We showed significant decrease in the expression of pro-angiogenic genes including HIF1-α, TGF-β, SOX2, Snail1, Snail2 and VEGFR in cells and also their secreted exosomes after treatment with DHA in normoxic and hypoxic conditions. Also the expression levels of tumor suppressor miRs including miR-101, miR-199, miR-342 were increased and the expression levels of oncomiRs including mir-382 and miR-21 were decreased after treatment with DHA in cells and exosomes. DHA can alter the expression of pro-angiogenic genes and microRNA contents in breast cancer cells and their derived-exosomes in favor of the inhibition of angiogenesis. Our data demonstrated new insight into DHA's anti-cancer action to target not only breast cancer cells but also their derived exosomes to suppress tumor angiogenesis. DHA has potent anti-angiogenic effects in breast cancer cells via down regulating expression of several angiogenic factors and also changing the contents of exosomes to convey anti-angiogenic effects of DHA to target cells. Unlabelled Image • Tumor-derived exosomes contain multiple pro-angiogenic cargoes. • DHA alters the contents of exosomes, besides cancer cells. • Exosomes can mediate DHA's anti-angiogenic action to target cells. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.