Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Front Immunol ; 15: 1353570, 2024.
Article in English | MEDLINE | ID: mdl-38646527

ABSTRACT

Despite significant advances in the development and refinement of immunotherapies administered to combat cancer over the past decades, a number of barriers continue to limit their efficacy. One significant clinical barrier is the inability to mount initial immune responses towards the tumor. As dendritic cells are central initiators of immune responses in the body, the elucidation of mechanisms that can be therapeutically leveraged to enhance their functions to drive anti-tumor immune responses is urgently needed. Here, we report that the dietary sugar L-fucose can be used to enhance the immunostimulatory activity of dendritic cells (DCs). L-fucose polarizes immature myeloid cells towards specific DC subsets, specifically cDC1 and moDC subsets. In vitro, L-fucose treatment enhances antigen uptake and processing of DCs. Furthermore, our data suggests that L-fucose-treated DCs increase stimulation of T cell populations. Consistent with our functional assays, single-cell RNA sequencing of intratumoral DCs from melanoma- and breast tumor-bearing mice confirmed transcriptional regulation and antigen processing as pathways that are significantly altered by dietary L-fucose. Together, this study provides the first evidence of the ability of L-fucose to bolster DC functionality and provides rational to further investigate how L-fucose can be used to leverage DC function in order to enhance current immunotherapy.


Subject(s)
Dendritic Cells , Fucose , Dendritic Cells/immunology , Dendritic Cells/metabolism , Animals , Mice , Fucose/metabolism , Antigen Presentation , Female , Mice, Inbred C57BL , Cell Polarity , Cell Line, Tumor , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Melanoma, Experimental/immunology , Lymphocyte Activation/immunology
2.
medRxiv ; 2023 Oct 16.
Article in English | MEDLINE | ID: mdl-37904926

ABSTRACT

Background: Currently there are no biomarkers to identify resistance to androgen-deprivation therapy (ADT) in men with hormone-naive prostate cancer. 5-hydroxymethylcytosines (5hmC) in the gene body are associated with gene activation and are critical for epigenomic regulation of cancer progression. Objective: To evaluate whether 5hmC signature in cell-free DNA (cfDNA) predicts early ADT resistance. Design Setting and Participants: Serial plasma samples from 55 prostate cancer patients receiving ADT were collected at three timepoints including baseline (prior to initiating ADT, N=55), 3-month (after initiating ADT, N=55), and disease progression (N=15) within 24 months or 24-month if no progression was detected (N=14). 20 of the 55 patients showed disease progression during the 24-month follow-up. The remaining 35 patients showed no progression in the same follow-up period. Outcome Measurements and Statistical Analysis: cfDNA (5-10ng) was used for selective chemical labeling (hMe-Seal) sequencing to map 5hmC abundance across the genome. Read counts in gene bodies were normalized with DESeq2. Differential methylation and gene set enrichment analyses were performed to identify the 5hmC-enriched genes and biological processes that were associated with disease progression. Kaplan-Meir analysis was utilized to determine the association of 5hmC signatures with progression-free survival. Results and Limitations: 5hmC-sequencing generated an average of 18.6 (range 6.03 to 42.43) million reads per sample with 98% (95-99%) mappable rate. Baseline sample comparisons identified significant 5hmC difference in 1,642 of 23,433 genes between 20 patients with progression and 35 patients without progression (false discovery rate, FDR<0.1). Patients with progression showed significant enrichments in multiple hallmark gene sets with androgen responses as the top enriched gene set (FDR=1.19E-13). Interestingly, this enrichment was driven by a subgroup of patients with disease progression featuring a significant 5hmC hypermethylation of the gene sets involving AR, FOXA1 and GRHL2. To quantify overall activities of these gene sets, we developed a gene set activity score algorithm using a mean value of log2 ratios of gene read counts in an entire gene set. We found that the activity scores in these gene sets were significantly higher in this subgroup of patients with progression than in the remaining patients regardless of the progression status. Furthermore, the high activity scores in these gene sets were associated with poor progression-free survival (p <0.05). Longitudinal analysis showed that activity scores in this subgroup with progression were significantly reduced after 3-month ADT but returned to high levels when the disease was progressed. Conclusions: 5hmC-sequencing in cfDNA identified a subgroup of prostate cancer patients with preexisting activation (5hmC hypermethylation) of gene sets involving AR, FOXA1 and GRHL2 before initiating ADT. Activity scores in these gene sets may serve as sensitive biomarkers to determine treatment resistance, monitor disease progression and potentially identify patients who would benefit from upfront treatment intensification. More studies are needed to validate this initial finding. Patient summary: There are no clinical tests to identify prostate cancer patients who will develop early resistance to androgen deprivation therapy within 24 months. In this study, we evaluated cell-free DNA epigenomic modification in blood and identified significant enrichment of 5-hydroxymethylation in androgen response genes in a subgroup of patients with treatment resistance. High level 5-hydroxylmethylation in these genes may serve as a discriminative biomarker to diagnose patients who are likely to experience early failure during androgen deprivation therapy.

3.
Pathol Res Pract ; 243: 154341, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36739754

ABSTRACT

Breast cancer is the most frequently diagnosed malignant tumor in women and a major public health concern. NRF2 axis is a cellular protector signaling pathway protecting both normal and cancer cells from oxidative damage. NRF2 is a transcription factor that binds to the gene promoters containing antioxidant response element-like sequences. In this report, differential expression of NRF2 signaling pathway elements, as well as the correlation of NRF2 pathway mRNAs with various clinicopathologic characteristics, including molecular subtypes, tumor grade, tumor stage, and methylation status, has been investigated in breast cancer using METABRIC and TCGA datasets. In the current report, our findings revealed the deregulation of several NRF2 signaling elements in breast cancer patients. Moreover, there were negative correlations between the methylation of NRF2 genes and mRNA expression. The expression of NRF2 genes significantly varied between different breast cancer subtypes. In conclusion, substantial deregulation of NRF2 signaling components suggests an important role of these genes in breast cancer. Because of the clear associations between mRNA expression and methylation status, DNA methylation could be one of the mechanisms that regulate the NRF2 pathway in breast cancer. Differential expression of Hippo genes among various breast cancer molecular subtypes suggests that NRF2 signaling may function differently in different subtypes of breast cancer. Our data also highlights an interesting link between NRF2 components' transcription and tumor grade/stage in breast cancer.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Prognosis , Transcriptome , Signal Transduction/genetics , RNA, Messenger/genetics
4.
BMC Cancer ; 22(1): 1282, 2022 Dec 07.
Article in English | MEDLINE | ID: mdl-36476410

ABSTRACT

Breast cancer is a complex disease exhibiting a great degree of heterogeneity due to different molecular subtypes. Notch signaling regulates the differentiation of breast epithelial cells during normal development and plays a crucial role in breast cancer progression through the abnormal expression of the Notch up-and down-stream effectors. To date, there are only a few patient-centered clinical studies using datasets characterizing the role of Notch signaling pathway regulators in breast cancer; thus, we investigate the role and functionality of these factors in different subtypes using publicly available databases containing records from large studies. High-throughput genomic data and clinical information extracted from TCGA were analyzed. We performed Kaplan-Meier survival and differential gene expression analyses using the HALLMARK_NOTCH_SIGNALING gene set. To determine if epigenetic regulation of the Notch regulators contributes to their expression, we analyzed methylation levels of these factors using the TCGA HumanMethylation450 Array data. Notch receptors and ligands expression is generally associated with the tumor subtype, grade, and stage. Furthermore, we showed gene expression levels of most Notch factors were associated with DNA methylation rate. Modulating the expression levels of Notch receptors and effectors can be a potential therapeutic approach for breast cancer. As we outline herein, elucidating the novel prognostic and regulatory roles of Notch implicate this pathway as an essential mediator controlling breast cancer progression.


Subject(s)
Breast Neoplasms , Transcriptome , Humans , Female , Prognosis , Breast Neoplasms/genetics , Epigenesis, Genetic , Gene Expression Profiling , Signal Transduction/genetics , Receptors, Notch/genetics
5.
Life Sci ; 309: 120975, 2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36126723

ABSTRACT

AIMS: Circular RNAs (circRNAs) are endogenous covalently closed non-coding RNAs produced by reverse splicing of linear RNA. These molecules are highly expressed in mammalian cells and show cell/tissue-specific expression patterns. They are also significantly dysregulated in various cancers and function as oncogenes or tumor suppressors. Emerging evidence reveals that circRNAs contribute to cancer progression via modulating different cell signaling pathways. Nevertheless, the functional significance of circRNAs in cell signaling pathways regulation is still largely elusive. Considering this, shedding light on the multi-pathway effects of circRNAs may improve our understanding of targeted cancer therapy. Here, we discuss how circRNAs regulate the major cell signaling pathways in human cancers. MATERIALS AND METHODS: We adopted a systematic search in PubMed using the following MeSH terms: circRNAs, non-coding RNAs, lncRNAs, exosomal circRNAs, cancer, and cell signaling. KEY FINDINGS: We discussed different roles of circRNAs during tumorigenesis in which circRNAs affect tumor development through activating or inactivating certain cell signaling pathways via molecular interactions using various signaling pathways. We also discussed how crosstalk between circRNAs and lncRNAs modulate tumorigenesis and provides a resource for the identification of cancer therapeutic targets. SIGNIFICANCE: We here elucidated how circRNAs can modulate different cell signaling pathways and play roles in cancer. This can broaden our horizons toward introducing promising prognostic, diagnostic, and therapeutic targets.


Subject(s)
Neoplasms , RNA, Long Noncoding , Animals , Humans , RNA, Circular/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Neoplasms/genetics , Neoplasms/diagnosis , Signal Transduction/genetics , Carcinogenesis , Mammals/genetics , Mammals/metabolism
6.
Ann N Y Acad Sci ; 1503(1): 5-22, 2021 11.
Article in English | MEDLINE | ID: mdl-34403146

ABSTRACT

Different cellular mechanisms contribute to osteocyte development. And while critical roles for members of the zinc finger protein SNAI family (SNAIs) have been discussed in cancer-related models, there are few reviews summarizing their importance for chondrocyte-to-osteocyte development. To help fill this gap, we review the roles of SNAIs in the development of mature osteocytes from chondrocytes, including the regulation of chondro- and osteogenesis through different signaling pathways and in programmed cell death. We also discuss how epigenetic factors-including DNA methylation, histone methylation and acetylation, and noncoding RNAs-contribute differently to both chondrocyte and osteocyte development. To better grasp the important roles of SNAIs in bone development, we also review genotype-phenotype correlations in different animal models. We end with comments about the possible importance of the SNAI family in cartilage/bone development and the potential applications for therapeutic goals.


Subject(s)
Cell Differentiation , Chondrocytes/cytology , Chondrocytes/metabolism , Gene Expression Regulation, Developmental , Osteocytes/cytology , Osteocytes/metabolism , Snail Family Transcription Factors/genetics , Animals , Bone Development/genetics , Cell Differentiation/genetics , Chondrogenesis/genetics , Epigenesis, Genetic , Humans , Osteogenesis/genetics , Signal Transduction , Snail Family Transcription Factors/metabolism
7.
Life Sci ; 277: 119488, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33862117

ABSTRACT

Glioblastoma is recognized as one of the leading causes of death worldwide. Although there have been considerable advancements in understanding the causative molecular mechanisms of this malignancy, effective therapeutic strategies are still in limited use. It has been revealed that non-coding RNAs (ncRNAs) play critical roles in glioblastoma development, while interactions between the regulatory molecules such as long ncRNAs (lncRNAs), microRNAs (miRNAs), transcribed pseudogenes, and circular RNAs (circRNAs) remain to be fully deciphered. Over the recent years, researchers have discovered a new category of RNA molecules called competing endogenous RNA (ceRNA). This kind of RNA can contribute to molecular interactions in the form of ceRNA networks (ceRNETs). Multiple lines of evidence have demonstrated that dysregulation of various ceRNA networks is involved in glioblastoma development. Therefore, gaining insights into these dysregulations might offer potential for the early diagnosis of glioblastoma patients and identification of efficient therapeutic targets. In this review, we provide an overview of recent discoveries on ceRNA networks and the involvement of dysregulated networks in posing limitations to temozolomide therapy. We also describe signaling pathways relevant to the progression of glioblastoma.


Subject(s)
Gene Regulatory Networks/genetics , Glioblastoma/genetics , RNA, Untranslated/genetics , Computational Biology/methods , Gene Expression Regulation, Neoplastic/genetics , Humans , MicroRNAs/genetics , RNA/genetics , RNA, Circular/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Untranslated/metabolism
8.
J Cell Physiol ; 236(9): 6200-6224, 2021 09.
Article in English | MEDLINE | ID: mdl-33559213

ABSTRACT

Posttranscriptional regulation is a mechanism for the cells to control gene regulation at the RNA level. In this process, RNA-binding proteins (RBPs) play central roles and orchestrate the function of RNA molecules in multiple steps. Accumulating evidence has shown that the aberrant regulation of RBPs makes  contributions to the initiation and progression of tumorigenesis via numerous mechanisms such as genetic changes, epigenetic alterations, and noncoding RNA-mediated regulations. In this article, we review the effects caused by RBPs and their functional diversity in the malignant transformation of cancer cells that occurs through the involvement of these proteins in various stages of RNA regulation including alternative splicing, stability, polyadenylation, localization, and translation. Besides this, we review the various interactions between RBPs and other crucial posttranscriptional regulators such as microRNAs and long noncoding RNAs in the pathogenesis of cancer. Finally, we discuss the potential approaches for targeting RBPs in human cancers.


Subject(s)
Carcinogenesis/metabolism , Neoplasms/metabolism , RNA-Binding Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Alternative Splicing/genetics , Humans , Neoplasms/pathology , Neoplasms/therapy , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
9.
Life Sci ; 264: 118719, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33159957

ABSTRACT

AIM: As a natural compound, docosahexaenoic acid (DHA) exerts anti-cancer and anti-angiogenesis functions through exosomes; however, little is known about the molecular mechanisms. MAIN METHODS: Breast cancer (BC) cells were treated with DHA (50 µM) and then tumor cell-derived exosomes (TDEs) were collected and characterized by electron microscopy, dynamic light scattering, and western blot analyses. By the time the cells were treated with DHA, RT-qPCR was used to investigate the expression of vascular endothelial growth factor (VEGF) and the selected pro- and anti-angiogenic microRNAs (miRNAs). The quantification of secreted VEGF protein was measured by enzyme-linked immunosorbent assay (ELISA). The effects of TDEs on endothelial cell angiogenesis were explored by transwell cell migration and in vitro vascular tube formation assays. KEY FINDINGS: DHA treatment caused a significant and time-dependent decrease in the expression and secretion of VEGF in/from BC cells. This also increased expression of anti-angiogenic miRNAs (i.e. miR-34a, miR-125b, miR-221, and miR-222) while decreased levels of pro-angiogenic miRNAs (i.e. miR-9, miR-17-5p, miR-19a, miR-126, miR-130a, miR-132, miR-296, and miR-378) in exosomes derived from DHA-treated BC cells, TDE (DHA+). While treatment with exosomes (100 µg/ml) obtained from untreated BC cells, TDE (DHA-), enhanced the expression of VEGF-A in human umbilical vein endothelial cells (HUVECs), incubation with DHA or TDE (DHA+) led to the significant decrease of VEGF-A transcript level in these cells. We indicated that the incubation with TDE (DHA+) could significantly decrease endothelial cell proliferation and migration and also the length and number of tubes made by HUVECs in comparison with endothelial cells incubated with exosomes obtained from untreated BC cells. SIGNIFICANCE: DHA alters angiogenesis by shifting the up-regulation of exosomal miRNA contents from pro-angiogenic to anti-angiogenic, resulting in the inhibition of endothelial cell angiogenesis. These data can help to figure out DHA's anti-cancer function, maybe its use in cancer therapy.


Subject(s)
Breast Neoplasms/pathology , Cell Movement/drug effects , Docosahexaenoic Acids/pharmacology , Exosomes/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Neovascularization, Physiologic/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Endocytosis/drug effects , Exosomes/drug effects , Exosomes/ultrastructure , Female , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Time Factors , Vascular Endothelial Growth Factor A/metabolism
10.
J Mol Endocrinol ; 66(2): R33-R55, 2021 02.
Article in English | MEDLINE | ID: mdl-33295879

ABSTRACT

Autoimmune thyroid disease (AITD) accounts for 90% of all thyroid diseases and affects 2-5% of the population with remarkable familial clustering. Among AITDs, Graves' disease (GD) is a complex disease affecting thyroid function. Over the last two decades, case-control studies using cutting-edge gene sequencing techniques have detected various susceptible loci that may predispose individuals to GD. It has been presumed that all likely associated genes, variants, and polymorphisms might be responsible for 75-80% of the heritability of GD. As a result, there are implications concerning the potential contribution of environmental and epigenetic factors in the pathogenesis of GD, including its initiation, progression, and development. Numerous review studies have summarized the contribution of genetic factors in GD until now, but there are still some key questions and notions that have not been discussed concerning the interplay of genetic, epigenetic, and immunological factors. With this in mind, this review discusses some newly-identified loci and their potential roles in the pathogenicity of GD. This may lead to the identification of new, promising therapeutic targets. Here, we emphasized principles, listed all the reported disease-associated genes and polymorphisms, and also summarized the current understanding of the epigenetic basis of GD.


Subject(s)
Epigenesis, Genetic , Genetic Predisposition to Disease , Graves Disease/genetics , Animals , Humans , Immune System/metabolism , T-Lymphocytes/metabolism , Thyroid Hormones/biosynthesis
11.
Eur J Med Genet ; 63(12): 104096, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33186761

ABSTRACT

Major facilitator superfamily domain-containing 2A (MFSD2A) is required for brain uptake of Docosahexaenoic acid and Lysophosphatidylcholine, both are essential for the normal neural development and function. Mutations in MFSD2A dysregulate the activity of this transporter in brain endothelial cells and can lead to microcephaly. In this study, we describe an 11-year-old male who is affected by autosomal recessive primary microcephaly 15. This patient also shows severe intellectual disability, recurrent respiratory and renal infections, low birth weight, and developmental delay. After doing clinical and neuroimaging evaluations, due to heterogeneity of neurogenetic disorders, no narrow clinical diagnosis was possible, therefore, we utilized targeted-exome sequencing to identify any causative genetic factors. This revealed a homozygous in-frame deletion (NM_001136493.1: c.241_243del; p.(Val81del)) in the MFSD2A gene as the most likely disease-susceptibility variant which was confirmed by Sanger sequencing. Neuroimaging revealed lateral ventricular asymmetry, corpus callosum hypoplasia, type B of cisterna magna, and widening of Sylvian fissures. All of these novel phenotypes are associated with autosomal recessive primary microcephaly-15 (MCPH15). According to the genotype-phenotype data, p.(Val81del) can be considered a likely pathogenic variant leading to non-lethal microcephaly. However, further cumulative data and molecular approaches are required to accurately identify genotype-phenotype correlations in MFSD2A.


Subject(s)
Developmental Disabilities/genetics , Microcephaly/genetics , Phenotype , Symporters/genetics , Cerebral Ventricles/diagnostic imaging , Child , Consanguinity , Corpus Callosum/diagnostic imaging , Developmental Disabilities/pathology , Gene Deletion , Genes, Recessive , Homozygote , Humans , Male , Microcephaly/pathology , Pedigree
12.
Arch Iran Med ; 23(10): 688-696, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33107310

ABSTRACT

BACKGROUND: Ankylosing spondylitis (AS; OMIM:106300) is a common complex inflammatory disease; in a previous study, we introduced a novel mutation in the RELN gene (OMIM: 600514) which was associated with AS. This study is designed to investigate the potential effect of RELN S2486G mutation on reelin secretion; additionally, we objected to evaluate the phospholipase A2 (PLA2G7) gene (OMIM: 601690) expression and platelet-activating factor-acetylhydrolase (PAF-AH) concentration as the downstream gene and the encoded protein. METHODS: The impact of the S2486G on reelin protein secretion was investigated in CHO-K1 and HEK-293T cells by constructing wild-type and mutant plasmids. Besides, the possible effect of the mutation on expression and concentration of PLA2G7 and PAF-AH in THP1 cells was assessed by quantitative real-time PCR (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA), respectively. The study was performed at Tarbiat Modares University, Tehran, Iran, from 2016 to 2018. RESULTS: Our results showed that S2486G not only causes a significant reduction in reelin secretion in both HEK-293T and CHO-K1 cells, but also it leads to a significant reduction in PLA2G7 gene expression (P value < 0.001) and protein level of PAF-AH in THP-1 cells (P value < 0.003). CONCLUSION: The S2486G mutation in RELN can alter inflammatory and, to some extent, osteogenesis pathways mediated by reduced secretion of reelin and also reduced expression of the PLA2G7 gene.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Nerve Tissue Proteins/genetics , Serine Endopeptidases/genetics , Spondylitis, Ankylosing/genetics , Animals , CHO Cells , Cricetulus , HEK293 Cells , Humans , Mutation , Reelin Protein , THP-1 Cells
13.
Mol Biol Rep ; 47(9): 7229-7251, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32789576

ABSTRACT

Lung cancer (LC) is among the leading causes of death all over the world and it is often diagnosed at advanced or metastatic stages. Exosomes, derived from circulating vesicles that are released from the multivesicular body, can be utilized for diagnosis and also the prognosis of LC at early stages. Exosomal proteins, RNAs, and DNAs can help to better discern the prognostic and diagnostic features of LC. To our knowledge, there are various reviews on LC and the contribution of exosomes, but none of them are about the exome techniques and also their efficiency in LC. To fill this gap, in this review, we summarize the recent investigations regarding isolation and also the characterization of exosomes of LC cells. Furthermore, we discuss the noncoding RNAs as biomarkers and their applications in the diagnosis and prognosis of LC. Finally, we compare the efficacy of exosome isolation methods to better fi + 6 + guring out feasible techniques.


Subject(s)
Biomarkers, Tumor , Cell-Free Nucleic Acids , Circulating Tumor DNA , Exosomes/metabolism , Lung Neoplasms/blood , Lung Neoplasms/diagnosis , Biomarkers, Tumor/blood , Biomarkers, Tumor/isolation & purification , Cell-Free Nucleic Acids/blood , Cell-Free Nucleic Acids/isolation & purification , Circulating Tumor DNA/blood , Circulating Tumor DNA/isolation & purification , Exosomes/pathology , Humans , Lung Neoplasms/pathology
14.
Cell Oncol (Dordr) ; 43(6): 961-988, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32495294

ABSTRACT

BACKGROUND: Gastric cancer (GC) is a major health issue in the Western world. Current clinical imperatives for this disease include the identification of more effective biomarkers to detect GC at early stages and enhance the prevention and treatment of metastatic and chemoresistant GC. The advent of non-coding RNAs (ncRNAs), particularly microRNAs (miRNAs) and long-non coding RNAs (lncRNAs), has led to a better understanding of the mechanisms by which GC cells acquire features of therapy resistance. ncRNAs play critical roles in normal physiology, but their dysregulation has been detected in a variety of cancers, including GC. A subset of ncRNAs is GC-specific, implying their potential application as biomarkers and/or therapeutic targets. Hence, evaluating the specific functions of ncRNAs will help to expand novel treatment options for GC. CONCLUSIONS: In this review, we summarize some of the well-known ncRNAs that play a role in the development and progression of GC. We also review the application of such ncRNAs in clinical diagnostics and trials as potential biomarkers. Obviously, a deeper understanding of the biology and function of ncRNAs underlying chemoresistance can broaden horizons toward the development of personalized therapy against GC.


Subject(s)
Drug Resistance, Neoplasm/genetics , RNA, Untranslated/genetics , Stomach Neoplasms/genetics , Animals , Cell Cycle/genetics , Epithelial-Mesenchymal Transition/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Untranslated/metabolism
15.
Eur J Hum Genet ; 28(6): 754-762, 2020 06.
Article in English | MEDLINE | ID: mdl-32001840

ABSTRACT

Ankylosing spondylitis (AS) is a common complex inflammatory disease; however, up to now distinct genes with monogenic pattern have not been reported for this disease. In the present study, we report a large Iranian family with several affected members with AS. DNAs of the three affected and two healthy cases were chosen for performing whole-exome sequencing (WES). After several filtering steps, candidate variants in the following genes were detected: RELN, DNMT1, TAF4ß, MUC16, DLG2, and FAM208. However, segregation analysis confirmed the association of only one variant, c.7456A>G; p.(Ser2486Gly) in the RELN gene with AS in this family. In addition, in silico predictions supported the probable pathogenicity of this variant. In this study, for the first time, we report a novel variant in the RELN gene, c.7456A>G; p.(Ser2486Gly), which completely co-segregates with AS. This association suggests potential insights into the pathophysiological bases of AS and it could broaden horizons toward new therapeutic strategies.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Mutation, Missense , Nerve Tissue Proteins/genetics , Serine Endopeptidases/genetics , Spondylitis, Ankylosing/genetics , Adult , Cell Adhesion Molecules, Neuronal/chemistry , Extracellular Matrix Proteins/chemistry , Female , Humans , Male , Middle Aged , Nerve Tissue Proteins/chemistry , Pedigree , Reelin Protein , Serine Endopeptidases/chemistry , Spondylitis, Ankylosing/pathology
16.
Mol Genet Genomic Med ; 8(3): e1118, 2020 03.
Article in English | MEDLINE | ID: mdl-31944631

ABSTRACT

BACKGROUND: Whole-exome sequencing (WES) has emerged as a successful diagnostic tool in molecular genetics laboratories worldwide. In this study, we aimed to find the potential genetic cause of skeletal disease, a heterogeneous disease, revealing the obvious short stature phenotype. In an Iranian family, we used solo-WES in a suspected patient to decipher the potential genetic cause(s). METHODS: A comprehensive clinical and genotyping examination was applied to suspect the disease of the patient. The solo clinical WES was exploited, and the derived data were filtered according to the standard pipelines. In order to validate the WES finding, the region harboring the candidate variant in the CTSK gene was amplified from genomic DNA and sequenced directly by Sanger sequencing. RESULTS: Sequence analysis revealed a rare novel nonsense variant, p.(Trp320*); c.905G>A, in the CTSK gene (NM_000396.3). In silico analysis shed light on the contribution of the variant to the pathogenicity of pycnodysostosis. This variant was confirmed by Sanger sequencing and further clinical examinations of the patient confirmed the disease. CONCLUSION: The present study shows a rare variant of the CTSK gene, which inherited as autosomal recessive, in an Iranian male patient with pycnodysostosis. Taken together, the novel nonsense CTSK variant meets the criteria of being likely pathogenic according to the American College of Medical Genetics and Genomics-the Association for Molecular Pathology (ACMG-AMP) variant interpretation guidelines.


Subject(s)
Cathepsin K/genetics , Pycnodysostosis/genetics , Adolescent , Codon, Nonsense , Genetic Testing , Humans , Male , Pycnodysostosis/pathology , Exome Sequencing
17.
J Clin Lab Anal ; 34(2): e23063, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31595567

ABSTRACT

BACKGROUND: Breast cancer (BC) is the most common malignancy among females with dismal quality of life in patients. It has been proven that epigenetic factors, especially microRNAs, are involved in breast carcinogenesis and progression. This study aimed to assess the expression and clinical performances of a five-microRNA signature (miR-127-3p, miR-133a-3p, miR-155-5p, miR-199b-5p, and miR-342-5p) in breast cancer and adjacent normal tissues to identify a potential biomarker for BC and investigate the relationship between their expression and clinicopathological features of BC patients as well. METHODS: In this case-control investigation, we recruited 50 pairs of tumor and matched non-tumor surgical specimens from patients diagnosed with BC. Expression levels of miR-127-3p, miR-133a-3p, miR-155-5p, miR-199b-5p, and miR-342-5p were measured in BC and adjacent normal tissues by RT-qPCR. RESULTS: We found that miR-127-3p, miR-133a-3p, miR-199b-5p, and miR-342-5p were significantly down-regulated, while miR-155-5p was significantly up-regulated in BC tumor tissues compared with the corresponding adjacent normal tissues. The decreased expression of miR-127-3p, miR-133a-3p, miR-342-5p, and up-regulation of miR-155-5p showed a significant correlation with disease stage. We also found a significant down-regulation of miR-127-3p, miR-199b-5p, and miR-342-5p compared in HER-2-negative patients. Our results indicated that miR-155-5p had a higher expression level in HER-2-positive patients. Receiver operating characteristic (ROC) curve analysis demonstrated that all these five microRNAs can serve as potential biomarkers to distinguish between tumor and non-tumor breast tissue samples. CONCLUSIONS: The present findings suggested that dysregulation of this five-miRNA signature might be considered as a promising and functional biomarker for BC diagnosis.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , MicroRNAs/genetics , Breast Neoplasms/surgery , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Receptor, ErbB-2/genetics
18.
Int J Mol Sci ; 20(18)2019 Sep 11.
Article in English | MEDLINE | ID: mdl-31514268

ABSTRACT

In the development of the skeleton, the long bones are arising from the process of endochondral ossification (EO) in which cartilage is replaced by bone. This complex process is regulated by various factors including genetic, epigenetic, and environmental elements. It is recognized that DNA methylation, higher-order chromatin structure, and post-translational modifications of histones regulate the EO. With emerging understanding, non-coding RNAs (ncRNAs) have been identified as another mode of EO regulation, which is consist of microRNAs (miRNAs or miRs) and long non-coding RNAs (lncRNAs). There is expanding experimental evidence to unlock the role of ncRNAs in the differentiation of cartilage cells, as well as the pathogenesis of several skeletal disorders including osteoarthritis. Cutting-edge technologies such as epigenome-wide association studies have been employed to reveal disease-specific patterns regarding ncRNAs. This opens a new avenue of our understanding of skeletal cell biology, and may also identify potential epigenetic-based biomarkers. In this review, we provide an updated overview of recent advances in the role of ncRNAs especially focus on miRNA and lncRNA in the development of bone from cartilage, as well as their roles in skeletal pathophysiology.


Subject(s)
Cartilage/growth & development , Cartilage/metabolism , RNA, Untranslated/genetics , Animals , Chondrocytes/cytology , Chondrocytes/metabolism , Epigenesis, Genetic , Growth Plate/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Untranslated/metabolism
19.
Cell Mol Biol (Noisy-le-grand) ; 65(6): 12-16, 2019 Jul 31.
Article in English | MEDLINE | ID: mdl-31472042

ABSTRACT

Reelin is a large extracellular glycoprotein secreted by Cajal-Retzius cells and has a main role during brain development, especially in neuronal migration. Reelin is comprised of N-terminal F-Spondin like domain, eight tandem repeats, and a highly conserved basic C-Terminal Region (CTR). The CTR main role in the secretion of Reelin has been investigated by advertently inducing deletion in whole or a part of this region; however, the role of CTR point mutations on the secretion of Reelin is shrouded in mystery. In this study, we performed experimental analyses on a sub-region of Human Reelin containing 5th and 6th repeats (R5-R6), a part of 8th repeat and the CTR which were amplified from cDNA of K562 and HEPG2(HepatocellularG2) cells and cloned into a mammalian expressional plasmid (pVP22/myc-His). Bioinformatics investigation was performed on the CTR at both level of nucleotide and amino acid as well as mutant type. Random mutagenesis by error-prone PCR method was utilized to induce mutation in the CTR. The secretion efficiency of recombinant wild-type and mutant Reelin constructs compared in cell lysate and supernatant isolated from the transiently transfected HEK 293T cells using 6XHistag ELISA method. In-vitro study demonstrated that the CTR alteration (S3440P) leads to impairment of Reelin secretion even after overexpression. Our results indicate that S3440P substitution is the highly conserved structure of the CTR has an important effect on Reelin secretion.


Subject(s)
Amino Acid Substitution , Cell Adhesion Molecules, Neuronal/chemistry , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Amino Acid Sequence , Conserved Sequence , Evolution, Molecular , HEK293 Cells , Humans , Mutation/genetics , Reelin Protein
20.
Cell Reprogram ; 21(5): 238-248, 2019 10.
Article in English | MEDLINE | ID: mdl-31369321

ABSTRACT

Generation of induced pluripotent stem cells (iPSCs) has been described as a powerful method to dedifferentiate the specialized cells to pluripotency. However, obtaining cancer-specific iPS cells (iPCs) encounters several barriers. The generation of iPCs provides valuable experimental platforms to mimic oncogenesis and offers potentials regarding drug screening. To overcome the difficulties regarding the iPC generation, we aimed at optimizing the generation of iPCs from glioblastoma multiform (GBM) cell lines and at understanding the potential barriers ahead of this process. The T731, T653, and mouse embryonic fibroblast cells were transduced by using retroviral plasmids encoding Oct4, Sox2, and Klf4. The cells were cultured on a layer of feeder cells for 14 days in iPS media and the obtained colonies were then picked and expanded to be evaluated for pluripotency markers by alkaline phosphatase staining, qRT-PCR, and Western blotting. Our findings confirmed resistance in cancer cells to achieve the pluripotency markers. In addition to designing technical tricks to obviate the barriers ahead of iPC generation, we suggested the small molecule PD98059 to enhance the efficiency of iPC generation from GBM cell lines. The resulting iPCs can further be used as a platform to study the mechanism of cancer formation and as a tool for drug screening for the treatment of patients with GBM.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Fibroblasts/cytology , Glioblastoma/pathology , Induced Pluripotent Stem Cells/cytology , Animals , Biomarkers/metabolism , Cell Culture Techniques , Cells, Cultured , Feeder Cells , Fibroblasts/metabolism , Glioblastoma/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Mice , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...