Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Biochem Biophys Res Commun ; 678: 135-143, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37634411

ABSTRACT

Hematological malignancies(HMs) are highly heterogeneous diseases with globally rising incidence. Despite major improvements in the management of HMs, conventional therapies have limited efficacy, and relapses with high mortality rates are still frequent. Cordycepin, a nucleoside analog extracted from Cordyceps species, represents a wide range of therapeutic effects, including anti-inflammatory, anti-tumor, and anti-metastatic activities. Cordycepin induces apoptosis in different subtypes of HMs by triggering adenosine receptors, death receptors, and several vital signaling pathways such as MAPK, ERK, PI3K, AKT, and GSK-3ß/ß-catenin. This review article summarizes the impact of utilizing cordycepin on HMs, and highlights its potential as a promising avenue for future cancer research based on evidence from in vitro and in vivo studies, as well as clinical trials.


Subject(s)
Hematologic Neoplasms , Humans , Glycogen Synthase Kinase 3 beta , Hematologic Neoplasms/drug therapy , Deoxyadenosines/pharmacology , Deoxyadenosines/therapeutic use , Apoptosis
2.
Biochem Biophys Res Commun ; 676: 13-20, 2023 10 08.
Article in English | MEDLINE | ID: mdl-37480688

ABSTRACT

INTRODUCTION: T-cell acute lymphoblastic leukemia is characterized by its fast progression rate and high complications. TRAIL can be used to trigger apoptosis in cancer cells with minimal effects on normal cells, but most of cancer cells develop resistance to this agent through various mechanisms. HDAC inhibitors like SAHA can be used to make cancer cells more susceptible to TRAIL-induced apoptosis. In this study, this hypothesis was tested on MOLT-4 cancer cell line. MATERIALS AND METHODS: The cells were divided into six groups including the control group, TRAIL 50 nM, TRAIL 100 nM, SAHA 2 µM, SAHA 2 µM + TRAIL 50 nM, and SAHA 2 µM + TRAIL 100 nM. Apoptosis was evaluated by flowcytometry after 24, 48 and 72 h. The expression levels of c-flip, DR4, DR5, CHOP, NF-κB, STAT3, Akt, and PI3K genes were investigated by quantitative real-time PCR. Data were analyzed using two-way variance analysis with Tukey's and Dunnett's multiple comparisons tests, and statistical significance was defined as having a p-value less than 0.05. RESULTS: Groups exposed to the combination of SAHA with TRAIL demonstrated the maximum apoptosis in MOLT-4 cells by increasing the expression of DR4, DR5, and CHOP and decreasing the expression of c-flip, STAT3, PI3k, Akt, and NF-kB genes. CONCLUSION: It can be concluded that SAHA increases the sensitivity of MOLT-4 cells to TRAIL-mediated apoptosis, which can be used as a strategy to overcome resistance to TRAIL in leukemic patients.


Subject(s)
Neoplasms , Proto-Oncogene Proteins c-akt , Humans , Apoptosis , Cell Line , Flow Cytometry , NF-kappa B , Phosphatidylinositol 3-Kinases
3.
Toxicol Ind Health ; 39(8): 451-463, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37303071

ABSTRACT

Exposure to numerous pollutants is prevalent in workplaces. Examination of combined exposure to different harmful physical factors and chemicals has offered new insights into toxicology in recent years. This study aimed to investigate the hematological alterations caused by exposure to noise and toluene. Twenty-four New Zealand white rabbits were exposed to 1000 ± 50 ppm toluene and/or 100 ± 5 dB noise for 14 consecutive days. Exposure to noise and toluene changed a number of parameters of white blood cells (WBC), red blood cells (RBC), and platelets on different days after the exposure. Simultaneous exposure to noise and toluene increased WBC, and exposure to noise and toluene alone decreased RBC. Exposure to noise and toluene alone increased basophile, monocyte, and neutrophil counts. The coefficient of variation of red blood cell distribution width (RDW-CV) and the standard deviation of red blood cell distribution width (RDW-SD) significantly increased after co-exposure to noise and toluene. Platelet levels increased in the noise-exposed and the co-exposed groups and decreased in the toluene-exposed group. Furthermore, co-exposure to noise and toluene induced dissimilar synergistic and antagonistic effects on the hematological indices. According to the results of this study, simultaneous exposure to toluene and noise can aggravate some hematotoxic effects compared to exposure to noise or toluene alone. The results also demonstrated the vital role of the modulatory mechanisms of the body in controlling the detrimental effects of stressors.


Subject(s)
Noise , Toluene , Rabbits , Animals , Toluene/toxicity
4.
Mol Biol Rep ; 49(1): 19-29, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34820749

ABSTRACT

BACKGROUND: The tumor necrosis factor (TNF)-related apoptosis-inducing ligand, TRAIL, an apoptosis-inducing cytokine, has attracted much attention in the treatment of cancer for its selective toxicity to malignant rather than normal cells. However, the apoptosis-inducing ability of TRAIL is weaker than expected primarily due to cancer cell resistance. As one of the dietary flavonoids, kaempferol, has been shown to be antiproliferative and might have a protective effect against TRAIL resistance, particularly for hematologic malignancies. METHODS AND RESULTS: Here, we studied the potential of kaempferol to enhance the TRAIL-induced cytotoxicity and apoptosis in human chronic myelogenous leukemia (CML) cell line K-562, as well as the expression of specific genes with impact on TRAIL signal regulation. Analysis of flowcytometry data showed that treatment with kaempferol did enhance sensitivity of CML cells to pro-apoptotic effects of anti-TRAIL antibody. Although the gene expression levels were heterogeneous, cFLIP, cIAP1 and cIAP2 expression were generally downregulated where co-treatment of kaempferol and TRAIL was employed and these effects appeared to be dose-dependent. We further demonstrated that the expression of death receptors 4 and 5 tended to increase subsequent to the combination treatment. CONCLUSIONS: Consequently, it is reasonable to conclude that sensitization of chronic leukemia cells to TRAIL by kaempferol in vitro should be considered as a way of focusing clinical attention on leukemia therapy.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Kaempferols/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Baculoviral IAP Repeat-Containing 3 Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitor of Apoptosis Proteins/genetics , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Ubiquitin-Protein Ligases/genetics
5.
Biotechnol Appl Biochem ; 69(2): 822-839, 2022 Apr.
Article in English | MEDLINE | ID: mdl-33786874

ABSTRACT

Mesenchymal stem cells (MSCs) are one of the most prominent cells in the bone marrow. MSCs can affect acute lymphocytic leukemia (ALL) cells under hypoxic conditions. With this aim, we used MOLT-4 cells as simulators of ALL cells cocultured with bone marrow mesenchymal stem cells (BMMSCs) under hypoxic conditions in vitro. Then, mRNA and protein expression of the MAT2A, PDK1, and HK2 genes were evaluated by real-time PCR and Western blot which was also followed by apoptosis measurement by a flow-cytometric method. Next, the methylation status of the target genes was investigated by MS-qPCR. Additionally, candidate gene expressions were examined after treatment with rapamycin using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. We found that the mRNA expression of the candidate genes was augmented under the hypoxic condition in which MAT2A was upregulated in cocultured cells compared to MOLT-4, while HK2 and PDK1 were downregulated. Moreover, we found an association between gene expression and promoter methylation levels of target genes. Besides, expressions of the candidate genes were decreased, while their methylation levels were promoted following treatment with rapamycin. Our results suggest an important role for the BMMSC in regulating the methylation of genes involved in cell survival in hypoxia conditions; however, we found no evidence to prove the MSCs' effect on directing malignant lymphoblastic cells to apoptosis.


Subject(s)
Mesenchymal Stem Cells , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Apoptosis/genetics , Bone Marrow Cells/metabolism , Cell Hypoxia/genetics , Humans , Hypoxia/metabolism , Mesenchymal Stem Cells/metabolism , Methionine Adenosyltransferase , Methylation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA, Messenger/metabolism , Sirolimus
6.
J Cell Physiol ; 236(6): 4097-4105, 2021 06.
Article in English | MEDLINE | ID: mdl-33184857

ABSTRACT

Multiple factors, including growth factors, are shown to be culprits of cancer outset and persistence. Among growth factors, insulin-like growth factors (IGFs) family are of more importance in the prognosis of blood malignancies. After binding to their corresponding receptor, IGFs initiate PI3K/AKT signaling pathway and increase the translation of intracellular proteins, such as cell division-related proteins. They also stimulate the transcription of cell division-related genes using the Ras-GTP pathway. In addition to organs such as the liver, IGFs are secreted by tumor cells and can cause growth and proliferation of self or tumor cells via autocrine and paracrine methods. Current studies indicate that decreasing the effects of IGF by blocking them, their receptors, or PI3K/AKT pathway using various drugs could help to suppress the division of tumor cells. Here, we delineate the role of the IGF family in hematologic malignancies and their potential mechanisms.


Subject(s)
Hematologic Neoplasms/metabolism , Somatomedins/metabolism , Animals , Antineoplastic Agents/therapeutic use , Gene Expression Regulation, Neoplastic , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Ligands , Receptors, Somatomedin/metabolism , Signal Transduction , Somatomedins/genetics , Somatomedins/therapeutic use
7.
J Cell Physiol ; 235(11): 8461-8471, 2020 11.
Article in English | MEDLINE | ID: mdl-32324267

ABSTRACT

MicroRNAs (miRNAs) characterized by small, noncoding RNAs have a fundamental role in the regulation of gene expression at the post-transcriptional level. Additionally, miRNAs have recently been identified as potential regulators of various genes involved in the pathogenesis of the autoimmune and inflammatory disease. So far, the interaction between miRNAs and T lymphocytes in the immune response as a new and significant topic has not been emphasized substantially. The role of miRNAs in different biological processes including apoptosis, immune checkpoints and the activation of immune cells is still unclear. Aberrant miRNA expression profile affects various aspects of T-cell function. Accordingly, in this literature review, we summarized the role of significant miRNAs in T-cell development processes. Consequently, we demonstrated precise mechanisms that candidate miRNAs interfere in Immune response mediated by different types of T cells. We believe that a good understanding of the interaction between miRNAs and immune response contributes to the new therapeutic strategies in relation to disease with an immunological origin.


Subject(s)
Cell Differentiation/genetics , Hemostasis/physiology , Immunity/genetics , T-Lymphocytes/immunology , Animals , Apoptosis/genetics , Humans , MicroRNAs/genetics
8.
Cell Biochem Funct ; 38(4): 334-346, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31833074

ABSTRACT

MicroRNAs (miRNAs) that are characterized by small, noncoding RNA have an essential role in the pathogenesis of human diseases, including cancer. Furthermore, miRNAs, as a new paradigm of epigenetic regulators, play an important role in normal development and cellular function. This literature review summarizes the recurrent mechanism of gene regulation through miRNAs and, consequently, the impact of regulated genes on different cellular processes, including proliferation, metastasis, prognosis, and apoptosis. Additionally, what is important to note is that the expression of miRNAs in various cancer cells is different, and miRNAs have various target genes in various cancers. Accordingly, a proper understanding of gene regulation by miRNAs contributes to new perspectives in miRNA-based therapeutic strategies. SIGNIFICANCE OF THE STUDY: MiRNAs are considered as a crucial regulator of gene expression. The genes also play an important role in the expression of miRNAs; as a result, there is a relationship between them. In recent years, targeted therapy with miRNAs has been a significant challenge. Studying the mechanisms through which miRNAs regulate various cancer cell processes, including apoptosis, proliferation, and metastasis, is very critical in the treatment of cancer through miRNAs. Definitely, a proper understanding of the impacts of aberrant expression of miRNAs on cancer cell processes leads to new therapeutic strategies in the targeted therapy with miRNAs.


Subject(s)
Apoptosis , Cell Proliferation , Gene Expression Regulation, Leukemic , MicroRNAs/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA, Neoplasm/metabolism , Humans , Neoplasm Metastasis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
9.
J Cell Physiol ; 234(9): 14783-14799, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30773635

ABSTRACT

Zinc finger E-box binding homeobox 2 (ZEB2) is a DNA-binding transcription factor, which is mainly involved in epithelial-to-mesenchymal transition (EMT). EMT is a conserved process during which mature and adherent epithelial-like state is converted into a mobile mesenchymal state. Emerging data indicate that ZEB2 plays a pivotal role in EMT-induced processes such as development, differentiation, and malignant mechanisms, for example, drug resistance, cancer stem cell-like traits, apoptosis, survival, cell cycle arrest, tumor recurrence, and metastasis. In this regard, the understanding of mentioned subjects in the development of normal and cancerous cells could be helpful in cancer complexity of diagnosis and therapy. In this study, we review recent findings about the biological properties of ZEB2 in healthy and cancerous states to find new approaches for cancer treatment.

10.
J Cell Physiol ; 234(8): 13233-13241, 2019 08.
Article in English | MEDLINE | ID: mdl-30589076

ABSTRACT

INTRODUCTION: Acute myeloid leukemia (AML) is the most common acute leukemia in adults. Conventional treatments are associated with cytotoxicity and systemic side effects. Hence, efforts in the field of cancer treatment are focused on finding the strategies which can specifically target the tumor cells without affecting the normal cells. TNF-related apoptosis-inducing ligand (TRAIL) is a biological cytokine which has the mentioned specificity, but the resistance of some cancer cells limits its use as a therapeutic strategy. Recent studies have shown that quercetin (QUR) can be used as a sensitizing agent alongside with TRAIL. The present study showed that QUR can increase the effect of TRAIL-induced cytotoxicity in KG-1 cells. MATERIALS AND METHODS: In this descriptive study, the IC50 dose for QUR in the KG-1 cell line was first determined by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay. Then, the cells were treated with TRAIL and QUR for 12, 24, and 48 hr. The rate of apoptosis was measured by Annexin V/propidium iodide assay. Also, the molecular evaluation of candidate genes was accomplished before and after the treatment. RESULTS: The results indicated that QUR could sensitize the KG-1 cells against the TRAIL-induced apoptosis. This outcome is achieved by increasing the messenger RNA expression levels of the death receptor genes and reducing the expression of antiapoptotic proteins, as well as decreasing the expression of the NF-κB subunit. CONCLUSION: Our findings suggest that QUR can sensitize the acute myeloid KG-1 cells against TRAIL. Moreover, the combinational therapy of these agents might promisingly improve the clinical efficacy of TRAIL in patients with AML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Leukemia, Myeloid, Acute/pathology , Quercetin/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Cell Line, Tumor , Drug Synergism , Humans
11.
J Cell Physiol ; 234(5): 6230-6243, 2019 05.
Article in English | MEDLINE | ID: mdl-30246336

ABSTRACT

BACKGROUND: Osteoblastic differentiation of mesenchymal stem cells (MSCs) is the principal stage during the restoration and regeneration of bone tissue. Epigenetic modifications such as DNA methylation play a key role in the differentiation process of stem cells. In this study, the methylation status of the promoter region of ZBTB16 and Twist1 genes and their role in controlling osteoblastic differentiation in MSCs was investigated during the osteoblastic differentiation of MSCs. METHODS: The MSCs were cultured under standard conditions and differentiated into the osteoblasts. We had three treatment groups including 5-azacytidine (methylation inhibitor), metformin (Twist-inhibitor), and procaine (Wnt/ß-catenin inhibitor) and a non-treated group (control). Methylation level of DNA in the promoter regions was monitored by methylation specific-quantitative polymerase chain reaction (PCR). Also, the mRNA levels of key genes in osteoblastic differentiation were measured using real-time PCR. RESULTS: ZBTB16 gene expression was upregulated, and promoter methylation was decreased. For Twist1 messenger RNA (mRNA) level decreased and promoter methylation increased during osteoblastic differentiation of MSCs. 5-Azacytidine caused a significant reduction in methylation and increased the mRNA expression of ZBTB16 and Twist1. Metformin repressed the Twist1 expression, and therefore osteoblastic differentiation was increased. On the opposite side, procaine could block the WNT/ß-catenin signaling pathway, as a consequence the gene expression of key genes involved in osteoblastic differentiation was declined. CONCLUSION: We found that methylation of DNA in the promoter region of ZBTB16 and Twist1 genes might be one of the main mechanisms that controlling the gene expression during osteoblastic differentiation of MSCs. Also, we could find an association between regulation of Twist1 and ZBTB16 genes and osteoblastic differentiation in MSCs by showing the relation between their expression and some key genes involved in osteoblastic differentiation. In addition, we found a connection between the Twist1 expression level and osteoblastic differentiation by using a Twist-inhibitor (metformin).


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation/genetics , Mesenchymal Stem Cells/cytology , Nuclear Proteins/genetics , Osteoblasts/cytology , Promyelocytic Leukemia Zinc Finger Protein/genetics , Twist-Related Protein 1/genetics , Cell Line , DNA Methylation/physiology , Humans , Mesenchymal Stem Cells/metabolism , Nuclear Proteins/biosynthesis , Osteoblasts/metabolism , Osteogenesis/genetics , Promyelocytic Leukemia Zinc Finger Protein/biosynthesis , Twist-Related Protein 1/biosynthesis
12.
J Cell Physiol ; 234(9): 15108-15122, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30652308

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent stem cells and show distinct features such as capability for self-renewal and differentiation into several lineages of cells including osteoblasts, chondrocytes, and adipocytes. In this study, the methylation status of the promoter region of zinc finger and BTB domain containing 16 (ZBTB16), twist-related protein 1(Twist1), de novo DNA methyltransferases 3A (DNMT3A), SRY-box 9 (Sox9), osteocalcin (OCN), and peroxisome proliferator-activated receptor γ2 (PPARγ2) genes and their messenger RNA (mRNA) expression levels were evaluated during the osteoblastic differentiation of MSCs (ODMSCs). We planned two experimental groups including zoledronic acid (ZA)-treated and nontreated cells (negative control) which both were differentiated into the osteoblasts. Methylation level of DNA in the promoter regions was assayed by methylation-specific-quantitative polymerase chain reaction (MS-qPCR), and mRNA levels of the target inhibitory/stimulatory genes during osteoblastic differentiation of MSCs were measured using real-time PCR. During the experimental induction of ODMSCs, the mRNA expression of the OCN gene was upregulated and methylation level of its promoter region was decreased. Moreover, Sox9 and PPARγ2 mRNA levels were attenuated and their promoter regions methylation levels were significantly augmented. However, the mRNA expression of the DNMT3A was not affected during the ODMSCs though its methylation rate was increased. In addition, ZA could enhance the expression of the ZBTB16 and decrease its promoter regions methylation and on the opposite side, it diminished mRNA expression of Sox9, Twist1, and PPARγ2 genes and increased their methylation rates. Intriguingly, ZA did not show a significant impact on gene expression and methylation levels the OCN and DNMT3A. We found that methylation of the promoter regions of Sox9, OCN, and PPARγ2 genes might be one of the main mechanisms adjusting the genes expression during the ODMSCs. Furthermore, we noticed that ZA can accelerate the MSCs differentiation to the osteoblast cells via two regulatory processes; suppression of osteoblastic differentiation inhibitor genes including Sox9, Twist1, and PPARγ2, and through promotion of the ZBTB16 expression.

13.
J Cell Physiol ; 233(5): 3729-3744, 2018 05.
Article in English | MEDLINE | ID: mdl-28771724

ABSTRACT

Breast cancer (BC) is the most prevalent cancer in women worldwide. Although extensive studies are ongoing concerning its intricate molecular mechanisms, development of novel therapies and more accurate diagnostic and prognostic approaches is still a challenge. Epithelial-mesenchymal transition (EMT) enables the invasion of metastatic cancer cells and has recently been highlighted in a Cancer Stem Cell (CSC) model of BC. Epigenetic events as well as miRNA expression are the master regulators of tumorigenesis and add a further layer to the complexity of BC pathogenesis. The miRNAs are related to epigenetic event and additionally affect epigenetic pathways. Recent evidence demonstrates that epigenetic mechanisms such as DNA methylation may control miRNA expression. Because each miRNA may regulate several target genes, dysregulation of miRNA caused by aberrant DNA methylation patterns of the locus may influence important downstream pathways. Furthermore, some miRNAs is believed to regulate important DNA methylator factors. Any disruption or modification of this intricate network can contribute to the disease process; thus, it is essential to understand these changes. Advancements in new sequencing technologies to detect DNA methylation patterns has provided the opportunity to determine differentially methylated regions (DMRs) of the miRNA locus and their effect on expression profiles to improve BC diagnosis and treatment. The current review examines the interplay of DNA methylation mechanisms and miRNA function in invasive tumorigenesis, specifically EMT and CSC of BC, to highlight its potential for advancements on BC etiology, diagnosis, and therapy.


Subject(s)
Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Promoter Regions, Genetic/genetics , Animals , Breast Neoplasms/diagnosis , Cell Transformation, Neoplastic/genetics , Humans , Neoplastic Stem Cells/pathology
14.
J Cell Physiol ; 233(10): 6470-6485, 2018 10.
Article in English | MEDLINE | ID: mdl-29741767

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL or Apo2L) is a member of the tumor necrosis factor (TNF) superfamily that induces apoptosis in different types of cancer cells via activation of caspase cascade. TRAIL interacts with its cognate receptors that placed on cancer cells surface, including TRAIL-R1 (death receptor 4, DR4), TRAIL-R2 (death receptor 5, DR5), TRAIL-R3 (decoy receptor 1, DcR1), TRAIL-R4 (decoy receptor 2, DcR2), and osteoprotegerin (OPG). Despite high apoptosis-inducing ability of TRAIL, various cancerous cells gain resistance to TRAIL gradually, and consequently TRAIL potential for apoptosis stimulation in these cells diminishes intensely. According to diverse ranges of examinations, intracellular anti-apoptotic proteins, such as cellular-FLICE inhibitory protein (c-FLIP), apoptosis inhibitors (IAPs), myeloid cell leukemia sequence 1 (MCL-1), BCL-2, BCL-XL, and survivin play key role in cancer cells resistance to TRAIL. These proteins attenuate cancer cells sensitivity to TRAIL via various functions, importantly through caspase cascade suppression. The c-FLIP avoids from caspase 8 activation by FADD via binding to caspase 8 cleavage of FADD. Moreover, it activates signaling pathways that involved in cancer cells survival and proliferation. Intriguingly, it appears that the down-regulation of intracellular anti-apoptotic proteins, particularly c-FLIP is effectiveness goal for TRAIL-resistant cancers therapy, because their up-regulation in association with poor prognosis has been observed in various types of TRAIL-resistant cancers. In this review, we tried to collect and examine investigations that researchers have been able to sensitize cancer cells to TRAIL through targeting of c-FLIP alone or with other intracellular anti-apoptotic proteins directly or indirectly. It seems that co-treatment of resistant cells by TRAIL with other therapeutic agents with the aim of intracellular anti-apoptotic proteins inhibition is hopeful and attractive approach to overcome various TRAIL-resistant cancers.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Neoplasms/drug therapy , TNF-Related Apoptosis-Inducing Ligand/genetics , Apoptosis/genetics , Cell Proliferation/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neoplasms/genetics , Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Survivin/genetics , bcl-X Protein/genetics
15.
J Cell Biochem ; 118(10): 3193-3204, 2017 10.
Article in English | MEDLINE | ID: mdl-28252217

ABSTRACT

CpG methylation of DNA takes part in a specific epigenetic memory that plays crucial roles in the differentiation and abnormality of the cells. The methylation pattern aberration of genomes is affected in three ways, namely DNA methyltransferase (DNMT), ten-eleven translocation (TET), and methyl-binding domain (MBD) proteins. Of these, TET enzymes have recently been demonstrated to be master modifier enzymes in the DNA methylation process. Additionally, recent studies emphasize that not only epigenetic phenomena play a role in controlling hypoxia pathway, but the hypoxia condition also triggers hypomethylation of genomes that may help with the expression of hypoxia pathway genes. In this study, we suggested that TET1 and TET2 could play a role in the demethylation of genomes under chemical hypoxia conditions. Herein, the evaluating methylation status and mRNA expression of mentioned genes were utilized through real-time PCR and methylation-specific PCR (MSP), respectively. Our results showed that TET1 and TET2 genes were overexpressed (P < 0.05) under chemical hypoxia conditions in Retinal Pigment Epithelial (RPE) cells, whereas the promoter methylation status of them were hypomethylated in the same condition. Therefore, chemical hypoxia not only causes overexpression of TET1 and TET2 but also could gradually do promoter demethylation of same genes. This is the first study to show the relationship between epigenetics and the expression of mentioned genes related to hypoxia pathways. Furthermore, it seems that these associations in RPE cells are subjected to chemical hypoxia as a mechanism that could play a crucial role in methylation pattern changes of hypoxia-related diseases such as cancer and ischemia. J. Cell. Biochem. 118: 3193-3204, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
DNA Methylation , DNA-Binding Proteins/biosynthesis , Epigenesis, Genetic , Mixed Function Oxygenases/biosynthesis , Promoter Regions, Genetic , Proto-Oncogene Proteins/biosynthesis , Retinal Pigment Epithelium/metabolism , Cell Hypoxia , Dioxygenases , Female , Humans , Male , Retinal Pigment Epithelium/cytology
16.
Cell Immunol ; 315: 1-10, 2017 05.
Article in English | MEDLINE | ID: mdl-28284487

ABSTRACT

Understanding the role of toll-like receptors (TLRs) in the immunomodulation potential, differentiation, migration, and survival of mesenchymal stem cells (MSCs) is absolutely vital to fully exploiting their MSC-based therapeutic potential. Furthermore, through recognition of exogenous or endogenous ligands produced upon injury, TLRs have been linked to allograft rejection and maintenance of chronic inflammatory diseases, including Crohn's disease, rheumatoid arthritis. Characterizing the effect of TLRs in biological control of MSCs fate and function could improve our knowledge about the MSC-based cell therapy and immunotherapy. In this paper, we outline the impacts of TLR activation and mechanisms on MSCs immunomodulatory functions, differentiation, migration, and survivability. Moreover, we indicate that the expression patterns of TLRs in MSCs from different sources.


Subject(s)
Cell Differentiation/physiology , Mesenchymal Stem Cells/physiology , Toll-Like Receptors/physiology , Adipogenesis/physiology , Animals , Apoptosis/physiology , Cell Lineage , Cell Movement/physiology , Chondrogenesis/physiology , Endosomes/physiology , Humans , Immune System/physiology , Inflammation/physiopathology , Mice , Organ Specificity , Osteogenesis/physiology , Pathogen-Associated Molecular Pattern Molecules
17.
Clin Transl Oncol ; 24(7): 1250-1261, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35076883

ABSTRACT

Drug resistance is the drug-effectiveness reduction in treatment and is a serious problem in oncology and infections. In oncology, drug resistance is a complicated process resulting from enhancing the function of a pump that transports drugs out of tumor cells, or acquiring mutations in drug target. Surprisingly, most drugs are very effective in the early stages, but the response to the drug wears off over time and resistance eventually develops. Drug resistance is caused by genetic and epigenetic changes that affect cancer cells and the tumor environment. The study of inherited changes in the phenotype without changes in the DNA sequence is called epigenetics. Because of reversible changes in epigenetics, they are an attractive target for therapy. Some of these epigenetic drugs are effective in treating cancers like acute myeloid leukemia (AML), which is characterized by the accumulation and proliferation of immature hematopoietic cells in the blood and bone marrow. In this article, we outlined the various contributing factors involved in resistance or sensitivity to epigenetic drugs in the treatment of AML.


Subject(s)
Leukemia, Myeloid, Acute , Bone Marrow/pathology , Epigenesis, Genetic , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mutation
18.
Pathol Res Pract ; 239: 154130, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36201929

ABSTRACT

BACKGROUND: However, advanced technologies have been developed in the treatment of various cancers, but the mortality rate from cancer is still very high. Drug resistance is a major problem for patients with cancer, which causes the treatment process to fail. In addition to inhibiting drug resistance, targeted therapy is also very important in treatment. MAIN BODY: Nowadays, miRNAs have gained increasing interest as they play a major role in both drug resistance and targeted therapy. MicroRNA (miRNA) is an important part of non-coding RNA that regulates gene expression at a post-transcriptional level. The prevailing studies about miRNA expression have been expanded into a variety of neoplasms. MiR-424 and miR-631 targets genes involved in various cellular processes and can participate in proliferation, differentiation, apoptosis, invasion, angiogenesis, and drug resistance and sensitivity. CONCLUSION: In this study, we focus on the role of miR-424 and miR-631 in many cancer types by displaying the potential target genes associated with each cancer, as well as briefly describing the clinical uses of miR-424 and miR-631 as a diagnostic and predictive tool in malignancies.


Subject(s)
MicroRNAs , Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Apoptosis , Drug Resistance , Gene Expression Regulation, Neoplastic/genetics
19.
Genes Dis ; 9(4): 849-867, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35685482

ABSTRACT

Blood disorders include a wide spectrum of blood-associated malignancies resulting from inherited or acquired defects. The ineffectiveness of existing therapies against blood disorders arises from different reasons, one of which is drug resistance, so different types of leukemia may show different responses to treatment. Leukemia occurs for a variety of genetic and acquired reasons, leading to uncontrolled proliferation in one or more cell lines. Regarding the genetic defects, oncogene signal transducer and activator of transcription (STAT) family transcription factor, especially STAT3, play an essential role in hematological disorders onset and progress upon mutations, dysfunction, or hyperactivity. Besides, microRNAs, as biological molecules, has been shown to play a dual role in either tumorigenesis and tumor suppression in various cancers. Besides, a strong association between STAT3 and miRNA has been reported. For example, miRNAs can regulate STAT3 via targeting its upstream mediators such as IL6, IL9, and JAKs or directly binding to the STAT3 gene. On the other hand, STAT3 can regulate miRNAs. In this review study, we aimed to determine the role of either microRNAs and STAT3 along with their effect on one another's activity and function in hematological malignancies.

20.
Asian Pac J Cancer Prev ; 22(6): 1975-1984, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-34181359

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the effect of mimic hypoxia on proliferation, the expression of significant miRNAs, and genes involved in drug resistance in MOLT-4 and KG1 cell lines. MATERIALS AND METHODS: The KG1 and MOLT-4 cell lines were cultured in RPMI 1640 medium supplemented with 20% FBS and 10% FBS respectively. The MTT test was used for determining  the optimum dose of CoCl2 for KG1 and MOLT-4 cell lines. Western blotting was used for the detection of HIF-1a protein and the confirmation of mimic hypoxia induced by CoCl2. For evaluating the effect of mimic hypoxia on proliferation of MOLT-4 and KG1 cell lines, cell counting was done using trypan blue at 24, 48, and 72 hours. Furthermore, the results obtained from cell counting were confirmed with the MTT test. Total RNA was extracted  using the RNX Plus solution kit according to the manufacturer's protocol. The expression of genes and miRNAs was evaluated with real time PCR. RESULTS: According to this study, mimic hypoxia induced by CoCl2 contributes to the overexpression of drug resistance related genes including MDR1, MRP1, FOXM1, BCL-xl genes, and the suppression of PUMA gene compared to the control group. The results also showed that mimic hypoxia condition leads to the up-regulation of miR-9 and down-regulation of miR-27a and miR-370. Additionally, our outcomes demonstrated that mimic hypoxia has an inhibitory effect on the proliferation of MOLT-4 and KG1 cell lines. CONCLUSION: Treatment with CoCl2 has an inhibitory effect on the proliferation of MOLT-4 and KG1 cell lines independent from real hypoxia. Additionally, mimic hypoxia has a substantial effect on the expression of  genes and miRNAs involved in drug resistance. Finally, we are still far away to discover the exact functional mechanisms of hypoxia on drug resistance but these evaluations can provide new perspectives into this field for the upcoming studies.


Subject(s)
Cell Hypoxia , Cobalt/pharmacology , MicroRNAs/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL