به جمع مشترکان مگیران بپیوندید!

تنها با پرداخت 70 هزارتومان حق اشتراک سالانه به متن مقالات دسترسی داشته باشید و 100 مقاله را بدون هزینه دیگری دریافت کنید.

برای پرداخت حق اشتراک اگر عضو هستید وارد شوید در غیر این صورت حساب کاربری جدید ایجاد کنید

عضویت

فهرست مطالب amir baghbanzadeh

  • Reza Dabbaghipour, Elham Ahmadi, Mona Entezam, Omid Rahbar Farzam, Amir Baghbanzadeh, Ali Saber Sichani, Nazila Jalilzadeh, Mahdi Jafarlou, Behzad Baradaran

    Scientific research over the past decades has proven the pivotal role of long non-coding RNAs (LncRNAs) in regulating gene expression. The immune responses are controlled through the interaction of pro-inflammatory (predominance of T helper 17 cells (Th17)) and anti-inflammatory cytokines excretion (predominance of Regulatory T cells (Treg)). Recent studies have marked the impact of many diverse LncRNAs on Treg/Th17 imbalances. Moreover, some of the roots and causes of human diseases can be associated with the alterations in the Th17/Treg ratio. In this review study, we overviewed the association between LncRNAs and Th17/Treg, with the potential of providing novel prognostic and diagnostic biomarkers and promising therapeutic targets in various diseases, particularly cancer.

    Keywords: Long non-coding RNA, Regulatory Tcells, Thelper 17cells}
  • Nazila Jalilzadeh, Neda Barzgar Barough, Mehrdad Karami, Amir Baghbanzadeh, Kobra Velaei*
    Introduction

     Understanding the key role of the tumor microenvironment in specifying molecular markers of breast cancer subtypes is of a high importance in diagnosis and treatment. Therefore, the possibility of interconversion of luminal states and their specific markers alteration under the control of tumor microenvironment (TME), particularly cancer-associated fibroblasts (CAFs) deserves to be further investigated.

    Methods

     To activate normal human fibroblasts, liquid overlay technique or nemosis was used and α-SMA protein expression, CAFs marker, in fibroblastic spheroids was measured by blotting. The luminal A, MCF-7, and luminal B, MDA-MB 361, cell lines were treated with normal and spheroidal/activated fibroblast conditioned medium for 48 hours. The morphological changes of both luminal A and B cells were evaluated by invert light microscopy and analyzed through the shape factor formula. Moreover, chemo-sensitivity, proliferation, and changes in ER-related and proliferative genes expression levels were assessed respectively via MTT assay, Ki67 expression Immunofluorescence assay, real time PCR and Annexin V-FITC techniques.

    Results

     Activated (spheroidal) fibroblasts, expressed αSMA marker two folds more than monolayer cultured fibroblasts. Our study indicated a significant increase in IC50 of both luminal A and B cell lines after being treated with conditioned medium particularly in treated group with spheroidal conditioned medium. Studying Morphological changes using shape factor formula demonstrated more aggressiveness with gaining mesenchymal features in both luminal A and B subtypes by increasing exposure time. Changes in the expression of Ki67 were observed following treatment with fibroblastic and spheroidal paracrine secretome. Driven Data from Ki67 assay supports the luminal A and B interconversion by elevated Ki67 expression in luminal A and lowered Ki67 expression in luminal B. Gene expression analysis revealed that anti-apoptotic Bcl2 gene expression in both luminal types treated with condition medium has been increased though there has seen no interchange in expression of ER-related and proliferative genes between luminal A (MCF7) and luminal B (MDA-MB361) subtypes, the results of Annexin V-FITC flow cytometry test indicated a decrease in the population of both early and late apoptotic cells in groups treated with both fibroblastic and spheroidal condition medium compared to of control group.

    Conclusion

     Under the paracrine influence of fibroblast cells, both luminal A (MCF7) and luminal B (MDA-MB) subtypes of breast cancer gained invasive, anti-apoptotic, and chemoresistance features which are mostly increased by activated(spheroidal) fibroblasts conditioned medium mimicking CAFs. There was no strong proof for interconversion of luminal A and luminal B which share more similarities among breast cancer molecular subtypes.

    Keywords: Subtype, Breast cancer, Tumor microenvironment, Cancer-associated fibroblast}
  • Farzaneh Bilan, Mohammad Amini, Mohammad Amin Doustvandi, Maryam Tohidast, Amir Baghbanzadeh, Seyed Samad Hosseini, Ahad Mokhtarzadeh *, Behzad Baradaran
    Introduction

    Gastric cancer (GC) is the third leading cause of cancer-related death worldwide. microRNAs are a group of regulatory non-coding RNAs that are involved in GC progression. miR-145 as a tumor suppressor and miR-21 as an oncomiR were shown to be dysregulated in many cancers including GC. This research aimed to enhance the expression of miR-145 while reducing the expression of miR- 21 and examine their impact on the proliferation, apoptosis, and migration of GC cells.

    Methods

    KATO III cells with high expression levels of miR-21-5p and low expression of miR-145-5p were selected. These cells were then transfected with either miR-145-5p mimics or anti-miR-21-5p, alone or in combination. Afterward, the cell survival rate was determined using the MTT assay, while apoptosis induction was investigated through V-FITC/PI and DAPI staining. Additionally, cell migration was examined using the wound healing assay, and cell cycle progression was analyzed through flow cytometry. Furthermore, gene expression levels were quantified utilizing the qRT-PCR technique.

    Results

    The study's findings indicated that the co-replacement of miR-145-5p and anti-miR-21- 5p led to a decrease in cell viability and the induction of apoptosis in GC cells. This was achieved via modulating the expression of Bax and Bcl-2, major cell survival regulators. Additionally, the combination therapy significantly increased sub-G1 cell cycle arrest and reduced cell migration by downregulating MMP-9 expression as an epithelial-mesenchymal transition marker. This study provides evidence for the therapeutic possibility of the combination of miR-145-5p and anti-miR- 21-5p and also suggests that they could inhibit cell proliferation by modulating the PTEN/AKT1 signaling pathway.

    Conclusion

    Our research revealed that utilizing miR-145-5p and anti-miR-21-5p together could be a promising therapeutic approach for treating GC.

    Keywords: Gastric cancer, miR-21-5p, miR-145-5p, Combination therapy, AKT signaling}
  • Monireh Khordadmehr*, Reyhaneh Matin, Behzad Baradaran*, Amir Baghbanzadeh, Farinaz Jigari-Asl, Farzad Katiraee

    MicroRNAs (miRNAs) are known as a group of short noncoding ribonucleic acids (ncRNAs). Mainly, they can manage gene expression at the posttranscriptional level in the essential biological and physiological functions. Significantly, more than 50% of the discovered miRNAs genes are placed in cancer‐related genomic regions, which can act as oncomiR or oncosuppressor. In this regard, growing evidence recently demonstrated the deregulation of miR-4800 in human cancers and non-cancerous diseases. However, little information is available on the biological roles of miR-4800 in cancer initiation, development, and progression. Here, we reviewed the targeting sites and biogenesis functions of the miR‐4800 family in physiological and pathological processes like human cancers, particularly with a particular focusing on the validated specific targets.

    Keywords: Cancer Progression, Microrna, Physiological Function, Therapeutic Aspect}
  • Amir Baghbanzadeh, Elham Baghbani, Khalil Hajiasgharzadeh, Saeed Noorolyai, Vahid Khaze, Behzad Mansoori, Masoud Shirmohamadi, Behzad Baradaran*, Ahad Mokhtarzadeh*
    Purpose

    microRNA‐193a‐5p is one of the well-known tumor suppressor miRNAs in the body but in many cases, its expression became reduced in patients suffering from gastric cancer (GC). The main purpose of this study was to restore the function of this miRNA in human GC cells and investigating the effects of enhanced expression of miR‐193a‐5p on proliferation, apoptosis, and migration of GC cells upon in vitro transfection.

    Methods

    The KATO III gastric cancer cells were treated with 100 nM of miR‐193a‐5p or negative control sequences. Following that, the MTT assay, flow cytometry assay, and wound-healing assay were applied to estimate the impacts of enhanced expression of this miRNA on the viability, apoptosis, and migration rate of the cells, respectively. Moreover, the total RNA was isolated and alterations in the mRNA expression ratio of migratory genes were measured by qRT-PCR techniques.

    Results

    The findings designated that enhanced expression of miR‐193a‐5p suppressed the migratory ability of the cells, but had no significant effects on cell survival or apoptosis of the transfected cells. In addition, this inhibitory function of miR‐193a‐5p on the migration rate of the KATO III cell line occurs with concurrent suppression of vimentin and MMP-9 gene expression.

    Conclusion

    It can be concluded that miR‐193a‐5p negatively influences the migratory ability of the cancerous cells and restoring its effects can be regarded as a promising target of future therapeutic interventions, especially for GC metastasis.

    Keywords: miRNA-193a‐5p, Gastric Cancer, Gene Therapy, Apoptosis, Migration Assay}
  • Somaiyeh Malekghasemi, Jafar Majidi, Amir Baghbanzadeh, Jalal Abdolalizadeh, Behzad Baradaran *, Leili Aghebati-Maleki

    Tumor microenvironment consists of malignant and non-malignant cells. The interaction of these dynamic and different cells is responsible for tumor progression at different levels. The non-malignant cells in TME contain cells such as tumor-associated macrophages (TAMs), cancer associated fibroblasts, pericytes, adipocytes, T cells, B cells, myeloid-derived suppressor cells (MDSCs), tumor-associated neutrophils (TANs), dendritic cells (DCs) and Vascular endothelial cells. TAMs are abundant in most human and murine cancers and their presence are associated with poor prognosis. The major event in tumor microenvironment is macrophage polarization into tumor-suppressive M1 or tumor-promoting M2 types. Although much evidence suggests that TAMS are primarily M2-like macrophages, the mechanism responsible for polarization into M1 and M2 macrophages remain unclear. TAM contributes cancer cell motility, invasion, metastases and angiogenesis. The relationship between TAM and tumor cells lead to used them as a diagnostic marker, therapeutic target and prognosis of cancer. This review presents the origin, polarization, role of TAMs in inflammation, metastasis, immune evasion and angiogenesis as well as they can be used as therapeutic target in variety of cancer cells. It is obvious that additional substantial and preclinical research is needed to support the effectiveness and applicability of this new and promising strategy for cancer treatment.

    Keywords: Tumor-associated macrophage (TAMs), Tumor microenvironment (TME), Therapeutic target, Malignant cells}
بدانید!
  • در این صفحه نام مورد نظر در اسامی نویسندگان مقالات جستجو می‌شود. ممکن است نتایج شامل مطالب نویسندگان هم نام و حتی در رشته‌های مختلف باشد.
  • همه مقالات ترجمه فارسی یا انگلیسی ندارند پس ممکن است مقالاتی باشند که نام نویسنده مورد نظر شما به صورت معادل فارسی یا انگلیسی آن درج شده باشد. در صفحه جستجوی پیشرفته می‌توانید همزمان نام فارسی و انگلیسی نویسنده را درج نمایید.
  • در صورتی که می‌خواهید جستجو را با شرایط متفاوت تکرار کنید به صفحه جستجوی پیشرفته مطالب نشریات مراجعه کنید.
درخواست پشتیبانی - گزارش اشکال