Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Semin Hematol ; 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38724414

ABSTRACT

microRNAs (miRNAs) are a class of small non-coding RNAs that play a crucial regulatory role in fundamental biological processes and have been implicated in various diseases, including cancer. The first evidence of the cancer-related function of miRNAs was discovered in chronic lymphocytic leukemia (CLL) in the early 2000s. Alterations in miRNA expression have since been shown to strongly influence the clinical course, prognosis, and response to treatment in patients with CLL. Therefore, the identification of specific miRNA alterations not only enhances our understanding of the molecular mechanisms underlying CLL but also holds promise for the development of novel diagnostic and therapeutic strategies. This review aims to provide a comprehensive summary of the current knowledge and recent insights into miRNA dysregulation in CLL, emphasizing its pivotal roles in disease progression, including the development of the lethal Richter syndrome, and to provide an update on the latest translational research in this field.

3.
Nat Rev Genet ; 25(3): 211-232, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37968332

ABSTRACT

Non-coding RNAs (ncRNAs) are a heterogeneous group of transcripts that, by definition, are not translated into proteins. Since their discovery, ncRNAs have emerged as important regulators of multiple biological functions across a range of cell types and tissues, and their dysregulation has been implicated in disease. Notably, much research has focused on the link between microRNAs (miRNAs) and human cancers, although other ncRNAs, such as long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), are also emerging as relevant contributors to human disease. In this Review, we summarize our current understanding of the roles of miRNAs, lncRNAs and circRNAs in cancer and other major human diseases, notably cardiovascular, neurological and infectious diseases. Further, we discuss the potential use of ncRNAs as biomarkers of disease and as therapeutic targets.


Subject(s)
MicroRNAs , Neoplasms , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Circular , RNA, Untranslated/genetics , Neoplasms/genetics , Neoplasms/therapy
5.
J Clin Invest ; 133(14)2023 07 17.
Article in English | MEDLINE | ID: mdl-37261908

ABSTRACT

Sepsis remains a leading cause of death for humans and currently has no pathogenesis-specific therapy. Hampered progress is partly due to a lack of insight into deep mechanistic processes. In the past decade, deciphering the functions of small noncoding miRNAs in sepsis pathogenesis became a dynamic research topic. To screen for new miRNA targets for sepsis therapeutics, we used samples for miRNA array analysis of PBMCs from patients with sepsis and control individuals, blood samples from 2 cohorts of patients with sepsis, and multiple animal models: mouse cecum ligation puncture-induced (CLP-induced) sepsis, mouse viral miRNA challenge, and baboon Gram+ and Gram- sepsis models. miR-93-5p met the criteria for a therapeutic target, as it was overexpressed in baboons that died early after induction of sepsis, was downregulated in patients who survived after sepsis, and correlated with negative clinical prognosticators for sepsis. Therapeutically, inhibition of miR-93-5p prolonged the overall survival of mice with CLP-induced sepsis, with a stronger effect in older mice. Mechanistically, anti-miR-93-5p therapy reduced inflammatory monocytes and increased circulating effector memory T cells, especially the CD4+ subset. AGO2 IP in miR-93-KO T cells identified important regulatory receptors, such as CD28, as direct miR-93-5p target genes. In conclusion, miR-93-5p is a potential therapeutic target in sepsis through the regulation of both innate and adaptive immunity, with possibly a greater benefit for elderly patients than for young patients.


Subject(s)
MicroRNAs , Sepsis , Humans , Mice , Animals , Aged , Antagomirs , MicroRNAs/genetics , Adaptive Immunity , Sepsis/pathology
6.
Proc Natl Acad Sci U S A ; 120(23): e2122053120, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37252969

ABSTRACT

The causes and consequences of abnormal biogenesis of extracellular vesicles (EVs) are not yet well understood in malignancies, including in breast cancers (BCs). Given the hormonal signaling dependence of estrogen receptor-positive (ER+) BC, we hypothesized that 17ß-estradiol (estrogen) might influence EV production and microRNA (miRNA) loading. We report that physiological doses of 17ß-estradiol promote EV secretion specifically from ER+ BC cells via inhibition of miR-149-5p, hindering its regulatory activity on SP1, a transcription factor that regulates the EV biogenesis factor nSMase2. Additionally, miR-149-5p downregulation promotes hnRNPA1 expression, responsible for the loading of let-7's miRNAs into EVs. In multiple patient cohorts, we observed increased levels of let-7a-5p and let-7d-5p in EVs derived from the blood of premenopausal ER+ BC patients, and elevated EV levels in patients with high BMI, both conditions associated with higher levels of 17ß-estradiol. In brief, we identified a unique estrogen-driven mechanism by which ER+ BC cells eliminate tumor suppressor miRNAs in EVs, with effects on modulating tumor-associated macrophages in the microenvironment.


Subject(s)
Breast Neoplasms , Extracellular Vesicles , MicroRNAs , Humans , Female , MicroRNAs/genetics , MicroRNAs/metabolism , Breast Neoplasms/pathology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estradiol/pharmacology , Estradiol/metabolism , Estrogens/metabolism , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Tumor Microenvironment
7.
Noncoding RNA ; 9(2)2023 Apr 13.
Article in English | MEDLINE | ID: mdl-37104009

ABSTRACT

Since the discovery of the first microRNAs (miRNAs, miRs), the understanding of miRNA biology has expanded substantially. miRNAs are involved and described as master regulators of the major hallmarks of cancer, including cell differentiation, proliferation, survival, the cell cycle, invasion, and metastasis. Experimental data indicate that cancer phenotypes can be modified by targeting miRNA expression, and because miRNAs act as tumor suppressors or oncogenes (oncomiRs), they have emerged as attractive tools and, more importantly, as a new class of targets for drug development in cancer therapeutics. With the use of miRNA mimics or molecules targeting miRNAs (i.e., small-molecule inhibitors such as anti-miRS), these therapeutics have shown promise in preclinical settings. Some miRNA-targeted therapeutics have been extended to clinical development, such as the mimic of miRNA-34 for treating cancer. Here, we discuss insights into the role of miRNAs and other non-coding RNAs in tumorigenesis and resistance and summarize some recent successful systemic delivery approaches and recent developments in miRNAs as targets for anticancer drug development. Furthermore, we provide a comprehensive overview of mimics and inhibitors that are in clinical trials and finally a list of clinical trials based on miRNAs.

8.
Cells ; 13(1)2023 12 30.
Article in English | MEDLINE | ID: mdl-38201290

ABSTRACT

MicroRNAs (miRNAs) are a type of non-coding RNA whose dysregulation is frequently associated with the onset and progression of human cancers. miR-142, an ultra-conserved miRNA with both active -3p and -5p mature strands and wide-ranging physiological targets, has been the subject of countless studies over the years. Due to its preferential expression in hematopoietic cells, miR-142 has been found to be associated with numerous types of lymphomas and leukemias. This review elucidates the multifaceted role of miR-142 in human physiology, its influence on hematopoiesis and hematopoietic cells, and its intriguing involvement in exosome-mediated miR-142 transport. Moreover, we offer a comprehensive exploration of the genetic and molecular landscape of the miR-142 genomic locus, highlighting its mutations and dysregulation within hematological malignancies. Finally, we discuss potential avenues for harnessing the therapeutic potential of miR-142 in the context of hematological malignancies.


Subject(s)
Exosomes , Hematologic Neoplasms , Leukemia , MicroRNAs , Humans , Hematologic Neoplasms/genetics , Hematologic Neoplasms/therapy , Leukemia/genetics , Leukemia/therapy , Exosomes/genetics , Genomics , MicroRNAs/genetics
9.
J Matern Fetal Neonatal Med ; 35(25): 8210-8216, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34470144

ABSTRACT

INTRODUCTION: The primary objective of the present study is to evaluate the effect of single-layer locked versus single-layer unlocked uterine closure techniques on the development of uterine niche. The secondary objective is to evaluate the effect of the ratio of lower uterine segment (LUS) to upper uterine segment (UUS) on the development of uterine niche, which was not previously investigated in literature but which the authors consider to be a major risk factor for the development of uterine niche. METHODS: Included in this randomized study were 194 patients who were admitted to the Department of Obstetrics and Gynecology at Health Sciences University Antalya Training and Research Hospital and who underwent cesarean section (CS) due to any reason between October 2017 and May 2018. Two different techniques were used in the closure of hysterotomy: Single-layer locked continuous suturing (Group 1) and single-layer unlocked continuous suturing (Group 2). During surgery, the thicknesses of the LUS and UUS were measured using a sterile scale prior to hysterotomy closure. The patients were evaluated for the development of uterine niche at postoperative six months by transvaginal ultrasound. RESULTS: Control transvaginal ultrasound performed at six months after surgery revealed uterine niches in 58 out of 194 patients (29.29%; 34 patients in Group 1 [34.3%] and 24 patients in Group 2 [25.3%]). No significant difference was noted in terms of the development of uterine niche between the two groups (p = .167). The mean LUS and UUS in patients without uterine niche development were 6.81 ± 1.26 mm and 9.38 ± 1.26 mm, whereas the mean LUS and UUS in patients with uterine niche development were 4.24 ± 1.15 mm and 9.21 ± 2.15, respectively (p = .001 and p = .236). The mean UUS/LUS ratio is 1.4 ± 0.16 among patients without uterine niche and 2.21 ± 0.31 in patients with uterine niche (p < .001). CONCLUSIONS: The present study found no statistically significant difference in niche size between the two groups. However, the study reports that the ratio of upper to lower uterine segment that was not previously investigated in literature is a major risk factor for the development of uterine niche.


Subject(s)
Cesarean Section , Cicatrix , Humans , Pregnancy , Female , Cesarean Section/adverse effects , Cesarean Section/methods , Cicatrix/etiology , Suture Techniques/adverse effects , Uterus/surgery , Uterus/pathology , Sutures
11.
RNA Biol ; 18(sup1): 416-429, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34387142

ABSTRACT

The human genome contains 481 ultraconserved regions (UCRs), which are genomic stretches of over 200 base pairs conserved among human, rat, and mouse. The majority of these regions are transcriptionally active (T-UCRs), and several have been found to be differentially expressed in tumours. Some T-UCRs have been functionally characterized, but of those few have been associated to breast cancer (BC). Using TCGA data, we found 302 T-UCRs related to clinical features in BC: 43% were associated with molecular subtypes, 36% with oestrogen-receptor positivity, 17% with HER2 expression, 12% with stage, and 10% with overall survival. The expression levels of 12 T-UCRs were further analysed in a cohort of 82 Brazilian BC patients using RT-qPCR. We found that uc.147 is high expressed in luminal A and B patients. For luminal A, a subtype usually associated with better prognosis, high uc.147 expression was associated with a poor prognosis and suggested as an independent prognostic factor. The lncRNA from uc.147 (lnc-uc.147) is located in the nucleus. Northern blotting results show that uc.147 is a 2,8 kb monoexonic trancript, and its sequence was confirmed by RACE. The silencing of uc.147 increases apoptosis, arrests cell cycle, and reduces cell viability and colony formation in BC cell lines. Additionally, we identifed 19 proteins that interact with lnc-uc.147 through mass spectrometry and demonstrated a high correlation of lnc-uc.147 with the neighbour gene expression and miR-18 and miR-190b. This is the first study to analyse the expression of all T-UCRs in BC and to functionally assess the lnc-uc.147.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Movement , Cell Proliferation , Female , Humans , Prognosis , Survival Rate , Tumor Cells, Cultured
13.
Mol Ther Nucleic Acids ; 23: 930-943, 2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33614241

ABSTRACT

Deregulation of noncoding RNAs, including microRNAs (miRs), is implicated in the pathogenesis of many human cancers, including breast cancer. Through extensive analysis of The Cancer Genome Atlas, we found that expression of miR-22-3p is markedly lower in triple-negative breast cancer (TNBC) than in normal breast tissue. The restoration of miR-22-3p expression led to significant inhibition of TNBC cell proliferation, colony formation, migration, and invasion. We demonstrated that miR-22-3p reduces eukaryotic elongation factor 2 kinase (eEF2K) expression by directly binding to the 3' untranslated region of eEF2K mRNA. Inhibition of EF2K expression recapitulated the effects of miR-22-3p on TNBC cell proliferation, motility, invasion, and suppression of phosphatidylinositol 3-kinase/Akt and Src signaling. Systemic administration of miR-22-3p in single-lipid nanoparticles significantly suppressed tumor growth in orthotopic MDA-MB-231 and MDA-MB-436 TNBC models. Evaluation of the tumor response, following miR-22-3p therapy in these models using a novel mathematical model factoring in various in vivo parameters, demonstrated that the therapy is highly effective against TNBC. These findings suggest that miR-22-3p functions as a tumor suppressor by targeting clinically significant oncogenic pathways and that miR-22-3p loss contributes to TNBC growth and progression. The restoration of miR-22-3p expression is a potential novel noncoding RNA-based therapy for TNBC.

14.
Gastroenterology ; 159(6): 2146-2162.e33, 2020 12.
Article in English | MEDLINE | ID: mdl-32805281

ABSTRACT

BACKGROUND & AIMS: Chromosomal instability (CIN) is a carcinogenesis event that promotes metastasis and resistance to therapy by unclear mechanisms. Expression of the colon cancer-associated transcript 2 gene (CCAT2), which encodes a long noncoding RNA (lncRNA), associates with CIN, but little is known about how CCAT2 lncRNA regulates this cancer enabling characteristic. METHODS: We performed cytogenetic analysis of colorectal cancer (CRC) cell lines (HCT116, KM12C/SM, and HT29) overexpressing CCAT2 and colon organoids from C57BL/6N mice with the CCAT2 transgene and without (controls). CRC cells were also analyzed by immunofluorescence microscopy, γ-H2AX, and senescence assays. CCAT2 transgene and control mice were given azoxymethane and dextran sulfate sodium to induce colon tumors. We performed gene expression array and mass spectrometry to detect downstream targets of CCAT2 lncRNA. We characterized interactions between CCAT2 with downstream proteins using MS2 pull-down, RNA immunoprecipitation, and selective 2'-hydroxyl acylation analyzed by primer extension analyses. Downstream proteins were overexpressed in CRC cells and analyzed for CIN. Gene expression levels were measured in CRC and non-tumor tissues from 5 cohorts, comprising more than 900 patients. RESULTS: High expression of CCAT2 induced CIN in CRC cell lines and increased resistance to 5-fluorouracil and oxaliplatin. Mice that expressed the CCAT2 transgene developed chromosome abnormalities, and colon organoids derived from crypt cells of these mice had a higher percentage of chromosome abnormalities compared with organoids from control mice. The transgenic mice given azoxymethane and dextran sulfate sodium developed more and larger colon polyps than control mice given these agents. Microarray analysis and mass spectrometry indicated that expression of CCAT2 increased expression of genes involved in ribosome biogenesis and protein synthesis. CCAT2 lncRNA interacted directly with and stabilized BOP1 ribosomal biogenesis factor (BOP1). CCAT2 also increased expression of MYC, which activated expression of BOP1. Overexpression of BOP1 in CRC cell lines resulted in chromosomal missegregation errors, and increased colony formation, and invasiveness, whereas BOP1 knockdown reduced viability. BOP1 promoted CIN by increasing the active form of aurora kinase B, which regulates chromosomal segregation. BOP1 was overexpressed in polyp tissues from CCAT2 transgenic mice compared with healthy tissue. CCAT2 lncRNA and BOP1 mRNA or protein were all increased in microsatellite stable tumors (characterized by CIN), but not in tumors with microsatellite instability compared with nontumor tissues. Increased levels of CCAT2 lncRNA and BOP1 mRNA correlated with each other and with shorter survival times of patients. CONCLUSIONS: We found that overexpression of CCAT2 in colon cells promotes CIN and carcinogenesis by stabilizing and inducing expression of BOP1 an activator of aurora kinase B. Strategies to target this pathway might be developed for treatment of patients with microsatellite stable colorectal tumors.


Subject(s)
Chromosomal Instability , Colorectal Neoplasms/genetics , Neoplasms, Experimental/genetics , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aurora Kinase B/metabolism , Azoxymethane/toxicity , Carcinogenesis/genetics , Cell Line, Tumor , Colon/cytology , Colon/pathology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/pathology , Cytogenetic Analysis , Dextrans/toxicity , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Male , Mice , Mice, Transgenic , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , Organoids , Primary Cell Culture , Proto-Oncogene Proteins c-myc/metabolism , RNA, Long Noncoding/genetics , RNA-Binding Proteins/metabolism , Signal Transduction/genetics
15.
Int J Mol Sci ; 21(9)2020 May 11.
Article in English | MEDLINE | ID: mdl-32403283

ABSTRACT

An increased focus is being placed on the tumorigenesis and contexture of tumor microenvironment in hematopoietic and solid tumors. Despite recent clinical revolutions in adoptive T-cell transfer approaches and immune checkpoint blockade, tumor microenvironment is a major obstacle to tumor regression in B-cell malignancies. A transcriptional alteration of coding and non-coding RNAs, such as microRNAs (miRNAs), has been widely demonstrated in the tumor microenvironment of B-cell malignancies. MiRNAs have been associated with different clinical-biological forms of B-cell malignancies and involved in the regulation of B lymphocyte development, maturation, and function, including B-cell activation and malignant transformation. Additionally, tumor-secreted extracellular vesicles regulate recipient cell functions in the tumor microenvironment to facilitate metastasis and progression by delivering miRNA contents to neighboring cells. Herein, we focus on the interplay between miRNAs and tumor microenvironment components in the different B-cell malignancies and its impact on diagnosis, proliferation, and involvement in treatment resistance.


Subject(s)
Gene Expression Regulation, Neoplastic , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , MicroRNAs/genetics , Tumor Microenvironment/genetics , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Proliferation/genetics , Extracellular Vesicles/genetics , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Lymphoma, Large B-Cell, Diffuse/diagnosis , Prognosis
16.
Gut ; 69(10): 1818-1831, 2020 10.
Article in English | MEDLINE | ID: mdl-31988194

ABSTRACT

OBJECTIVE: To investigate the function of a novel primate-specific long non-coding RNA (lncRNA), named FLANC, based on its genomic location (co-localised with a pyknon motif), and to characterise its potential as a biomarker and therapeutic target. DESIGN: FLANC expression was analysed in 349 tumours from four cohorts and correlated to clinical data. In a series of multiple in vitro and in vivo models and molecular analyses, we characterised the fundamental biological roles of this lncRNA. We further explored the therapeutic potential of targeting FLANC in a mouse model of colorectal cancer (CRC) metastases. RESULTS: FLANC, a primate-specific lncRNA feebly expressed in normal colon cells, was significantly upregulated in cancer cells compared with normal colon samples in two independent cohorts. High levels of FLANC were associated with poor survival in two additional independent CRC patient cohorts. Both in vitro and in vivo experiments demonstrated that the modulation of FLANC expression influenced cellular growth, apoptosis, migration, angiogenesis and metastases formation ability of CRC cells. In vivo pharmacological targeting of FLANC by administration of 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine nanoparticles loaded with a specific small interfering RNA, induced significant decrease in metastases, without evident tissue toxicity or pro-inflammatory effects. Mechanistically, FLANC upregulated and prolonged the half-life of phosphorylated STAT3, inducing the overexpression of VEGFA, a key regulator of angiogenesis. CONCLUSIONS: Based on our findings, we discovered, FLANC as a novel primate-specific lncRNA that is highly upregulated in CRC cells and regulates metastases formation. Targeting primate-specific transcripts such as FLANC may represent a novel and low toxic therapeutic strategy for the treatment of patients.


Subject(s)
Carcinogenesis , Cell Proliferation , Colorectal Neoplasms , Neovascularization, Pathologic , RNA, Long Noncoding , STAT3 Transcription Factor/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/therapy , Drug Discovery , Gene Expression Regulation, Neoplastic , Genetic Markers , Genetic Therapy , Humans , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Pharmacogenomic Testing , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Vascular Endothelial Growth Factor A/metabolism
18.
JCI Insight ; 5(1)2020 01 16.
Article in English | MEDLINE | ID: mdl-31941838

ABSTRACT

Myelofibrosis (MF) is a myeloproliferative neoplasm characterized by cytopenia and extramedullary hematopoiesis, resulting in splenomegaly. Multiple pathological mechanisms (e.g., circulating cytokines and genetic alterations, such as JAKV617F mutation) have been implicated in the etiology of MF, but the molecular mechanism causing resistance to JAK2V617F inhibitor therapy remains unknown. Among MF patients who were treated with the JAK inhibitor ruxolitinib, we compared noncoding RNA profiles of ruxolitinib therapy responders versus nonresponders and found miR-543 was significantly upregulated in nonresponders. We validated these findings by reverse transcription-quantitative PCR. in this same cohort, in 2 additional independent MF patient cohorts from the United States and Romania, and in a JAK2V617F mouse model of MF. Both in vitro and in vivo models were used to determine the underlying molecular mechanism of miR-543 in MF. Here, we demonstrate that miR-543 targets the dioxygenases ten-eleven translocation 1 (TET1) and 2 (TET2) in patients and in vitro, causing increased levels of global 5-methylcytosine, while decreasing the acetylation of histone 3, STAT3, and tumor protein p53. Mechanistically, we found that activation of STAT3 by JAKs epigenetically controls miR-543 expression via binding the promoter region of miR-543. Furthermore, miR-543 upregulation promotes the expression of genes related to drug metabolism, including CYP3A4, which is involved in ruxolitinib metabolism. Our findings suggest miR-543 as a potentially novel biomarker for the prognosis of MF patients with a high risk of treatment resistance and as a potentially new target for the development of new treatment options.


Subject(s)
DNA-Binding Proteins/drug effects , Epigenesis, Genetic/drug effects , MicroRNAs/metabolism , MicroRNAs/pharmacology , Primary Myelofibrosis/drug therapy , Proto-Oncogene Proteins/drug effects , Animals , Cytokines/metabolism , DNA-Binding Proteins/genetics , Dioxygenases , Disease Models, Animal , Histones , Humans , Janus Kinase Inhibitors/therapeutic use , Janus Kinases/metabolism , Mice , MicroRNAs/genetics , Mixed Function Oxygenases , Mutation , Myeloproliferative Disorders , Nitriles , Primary Myelofibrosis/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/genetics , Pyrazoles/therapeutic use , Pyrimidines , STAT3 Transcription Factor , Transcriptome , United States
19.
Cell Signal ; 68: 109539, 2020 04.
Article in English | MEDLINE | ID: mdl-31935430

ABSTRACT

High-grade serous ovarian carcinoma (HGSOC) is the most lethal gynecologic cancer. Emerging evidence suggests that tumor-associated macrophages (TAMs) play an immunosuppressive role in the tumor microenvironment and promote tumor growth, angiogenesis, and metastasis in ovarian cancer. Therefore, targeting TAMs in patients with ovarian cancer is an appealing strategy; however, all trials to date have failed. To improve the efficacy of this approach, we sought to elucidate the underlying mechanisms of the role of TAMs in ovarian cancer. We found that the developmental transcription factor GATA3 was highly expressed in HGSOC cell lines but not in the fallopian tube, which is the main origin of HGSOC. GATA3 expression was associated with poor prognosis in HGSOC patients (P < .05) and was found to promote proliferation and migration in HGSOC cell lines. GATA3 was released abundantly from TAM cells via exosomes and contributed to tumor growth in the tumor microenvironment. Moreover, GATA3 acted as a regulator for macrophage polarization and interactions between TAMs and HGSOC to support proliferation, motility, and cisplatin chemoresistance in mutant TP53 HGSOC cell lines. Furthermore, GATA3 played a critical role in the interactions between TAMs and mutant TP53 HGSOC to promote angiogenesis and epithelial-mesenchymal transition with epigenetic regulation. Targeting GATA3 using GATA3siRNA in TAMs impeded GATA3-driven proliferation, migration, cisplatin chemoresistance, and angiogenesis in mutant TP53 HGSOC cell lines. Our findings indicate that GATA3 plays a novel role in immunoediting of HGSOC and demonstrate that GATA3 may serve as a prognostic marker for HGSOC and a promising target in the treatment of HGSOC.


Subject(s)
GATA3 Transcription Factor/metabolism , Neoplasms, Cystic, Mucinous, and Serous/metabolism , Neoplasms, Cystic, Mucinous, and Serous/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Tumor-Associated Macrophages/metabolism , Apoptosis/genetics , Cell Communication/genetics , Cell Line, Tumor , Cell Movement , Cell Polarity/genetics , Endometrial Neoplasms/pathology , Endothelial Cells/pathology , Epigenesis, Genetic , Epithelial-Mesenchymal Transition/genetics , Exosomes/metabolism , Exosomes/ultrastructure , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Genome, Human , Humans , Matrix Metalloproteinase 9/metabolism , Mutation/genetics , Neoplasm Grading , Neoplasm Proteins/metabolism , Neoplasms, Cystic, Mucinous, and Serous/genetics , Neovascularization, Pathologic/genetics , Ovarian Neoplasms/genetics , Phosphorylation , RNA Splice Sites/genetics , Tumor Microenvironment/genetics , Tumor Suppressor Protein p53/genetics , Tumor-Associated Macrophages/pathology
20.
Oncogene ; 38(33): 6095-6108, 2019 08.
Article in English | MEDLINE | ID: mdl-31289363

ABSTRACT

Current anti-angiogenic therapy for cancer is based mainly on inhibition of the vascular endothelial growth factor pathway. However, due to the transient and only modest benefit from such therapy, additional approaches are needed. Deregulation of microRNAs (miRNAs) has been demonstrated to be involved in tumor angiogenesis and offers opportunities for a new therapeutic approach. However, effective miRNA-delivery systems are needed for such approaches to be successful. In this study, miRNA profiling of patient data sets, along with in vitro and in vivo experiments, revealed that miR-204-5p could promote angiogenesis in ovarian tumors through THBS1. By binding with scavenger receptor class B type 1 (SCARB1), reconstituted high-density lipoprotein-nanoparticles (rHDL-NPs) were effective in delivering miR-204-5p inhibitor (miR-204-5p-inh) to tumor sites to suppress tumor growth. These results offer a new understanding of miR-204-5p in regulating tumor angiogenesis.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/genetics , MicroRNAs , Neovascularization, Pathologic/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Angiogenesis Inhibitors/pharmacology , Animals , Carcinoma, Ovarian Epithelial/blood supply , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Mice , Mice, Nude , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Molecular Targeted Therapy/methods , Neovascularization, Pathologic/drug therapy , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/pathology , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...