1
|
Li K, Diakite D, Austin J, Lee J, Lantvit DD, Murphy BT, Burdette JE. The Flavonoid Baicalein Negatively Regulates Progesterone Target Genes in the Uterus in Vivo. JOURNAL OF NATURAL PRODUCTS 2022; 85:237-247. [PMID: 34935393 PMCID: PMC9164990 DOI: 10.1021/acs.jnatprod.1c01008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Baicalein is a flavonoid extracted from the root of Scutellaria baicalensis (Chinese skullcap) and is consumed as part of this botanical dietary supplement to reduce oxidative stress, pain, and inflammation. We previously reported that baicalein can also modify receptor signaling through the progesterone receptor (PR) and glucocorticoid receptor (GR) in vitro, which is interesting due to the well-established roles of both PR and GR in reducing inflammation. To understand the effects of baicalein on PR and GR signaling in vivo in the uterus, ovariectomized CD-1 mice were treated with DMSO, progesterone (P4), baicalein, P4 with baicalein, and P4 with RU486, a PR antagonist, for a week. The uteri were collected for histology and RNA sequencing. Our results showed that baicalein attenuated the antiproliferative effect of P4 on luminal epithelium as well as on the PR target genes HAND2 and ZBTB16. Baicalein did not change levels of PR or GR RNA or protein in the uterus. RNA sequencing data indicated that many transcripts significantly altered by baicalein were regulated in the opposite direction by P4. Similarly, a large portion of GO/KEGG terms and GSEA gene sets were altered in the opposite direction by baicalein as compared to P4 treatment. Treatment of baicalein did not change body weight, organ weight, or blood glucose level. In summary, baicalein functioned as a PR antagonist in vivo and therefore may oppose P4 action under certain conditions such as uterine hyperplasia, fibroids, and uterine cancers.
Collapse
Affiliation(s)
- Kailiang Li
- Department of Pharmaceutical Sciences, Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Djeneba Diakite
- Department of Pharmaceutical Sciences, Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Julia Austin
- Department of Pharmaceutical Sciences, Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Jeongho Lee
- Department of Pharmaceutical Sciences, Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Daniel D. Lantvit
- Department of Pharmaceutical Sciences, Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Brian T. Murphy
- Department of Pharmaceutical Sciences, Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Joanna E. Burdette
- Department of Pharmaceutical Sciences, Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60607, USA
| |
Collapse
|
2
|
Chuang TD, Quintanilla D, Boos D, Khorram O. Long Noncoding RNA MIAT Modulates the Extracellular Matrix Deposition in Leiomyomas by Sponging MiR-29 Family. Endocrinology 2021; 162:6365958. [PMID: 34491311 PMCID: PMC8459448 DOI: 10.1210/endocr/bqab186] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Indexed: 01/01/2023]
Abstract
The objective of this study was to determine the expression and functional role of a long noncoding RNA (lncRNA) MIAT (myocardial infarction-associated transcript) in leiomyoma pathogenesis. Leiomyoma compared with myometrium (n = 66) expressed significantly more MIAT that was independent of race/ethnicity and menstrual cycle phase but dependent on MED12 (mediator complex subunit 12) mutation status. Leiomyomas bearing the MED12 mutation expressed higher levels of MIAT and lower levels of microRNA 29 family (miR-29a, -b, and -c) compared with MED12 wild-type leiomyomas. Using luciferase reporter activity and RNA immunoprecipitation analysis, MIAT was shown to sponge the miR-29 family. In a 3-dimensional spheroid culture system, transient transfection of MIAT siRNA in leiomyoma smooth muscle cell (LSMC) spheroids resulted in upregulation of miR-29 family and downregulation of miR-29 targets, collagen type I (COL1A1), collagen type III (COL3A1), and TGF-β3 (transforming growth factor β-3). Treatment of LSMC spheroids with TGF-β3 induced COL1A1, COL3A1, and MIAT levels, but repressed miR-29 family expression. Knockdown of MIAT in LSMC spheroids blocked the effects of TGF-β3 on the induction of COL1A1 and COL3A1 expression. Collectively, these results underscore the physiological significance of MIAT in extracellular matrix accumulation in leiomyoma.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of Ob/Gyn Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, CA 90502, USA
| | - Derek Quintanilla
- Department of Ob/Gyn Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, CA 90502, USA
| | - Drake Boos
- Department of Ob/Gyn Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, CA 90502, USA
| | - Omid Khorram
- Department of Ob/Gyn Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, CA 90502, USA
- Correspondence: Omid Khorram, MD, PhD, Department of Ob/Gyn, Harbor-UCLA Medical Center, 1124 W. Carson St., Box 467, Torrance, CA 90502, USA.
| |
Collapse
|
3
|
Mesa AM, Mao J, Medrano TI, Bivens NJ, Jurkevich A, Tuteja G, Cooke PS, Rosenfeld CS. Spatial Transcriptomics analysis of uterine gene expression in enhancer of Zeste homolog 2 (Ezh2) conditional knockout mice. Biol Reprod 2021; 105:1126-1139. [PMID: 34344022 DOI: 10.1093/biolre/ioab147] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/12/2021] [Indexed: 12/16/2022] Open
Abstract
Histone proteins undergo various modifications that alter chromatin structure, including addition of methyl groups. Enhancer of homolog 2 (EZH2), is a histone methyltransferase that methylates lysine residue 27, and thereby, suppresses gene expression. EZH2 plays integral role in the uterus and other reproductive organs. We have previously shown that conditional deletion of uterine EZH2 results in increased proliferation of luminal and glandular epithelial cells, and RNAseq analyses reveal several uterine transcriptomic changes in Ezh2 conditional (c) knockout (KO) mice that can affect estrogen signaling pathways. To pinpoint the origin of such gene expression changes, we used the recently developed spatial transcriptomics (ST) method with the hypotheses that Ezh2cKO mice would predominantly demonstrate changes in epithelial cells and/or ablation of this gene would disrupt normal epithelial/stromal gene expression patterns. Uteri were collected from ovariectomized adult WT and Ezh2cKO mice and analyzed by ST. Asb4, Cxcl14, Dio2, and Igfbp5 were increased, Sult1d1, Mt3, and Lcn2 were reduced in Ezh2cKO uterine epithelium vs. WT epithelium. For Ezh2cKO uterine stroma, differentially expressed key hub genes included Cald1, Fbln1, Myh11, Acta2, and Tagln. Conditional loss of uterine Ezh2 also appears to shift the balance of gene expression profiles in epithelial vs. stromal tissue toward uterine epithelial cell and gland development and proliferation, consistent with uterine gland hyperplasia in these mice. Current findings provide further insight into how EZH2 may selectively affect uterine epithelial and stromal compartments. Additionally, these transcriptome data might provide the mechanistic understanding and valuable biomarkers for human endometrial disorders with epigenetic underpinnings.
Collapse
Affiliation(s)
- Ana M Mesa
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA.,Grupo de Investigación en Agrociencias, Biodiversidad y Territorio - GAMMA, Facultad de Ciencias Agrarias, Universidad de Antioquia UdeA, Calle 70 N° 52-21, Medellín, Colombia
| | - Jiude Mao
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA.,Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Theresa I Medrano
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Nathan J Bivens
- Genomics Technology, University of Missouri, Columbia, MO 65211, USA
| | - Alexander Jurkevich
- Advanced Light Microscopy Core Facility, University of Missouri, Columbia, MO 65211, USA
| | - Geetu Tuteja
- Genetics, Development and Cell Biology, Iowa State University, Ames, IA 50011, USA
| | - Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Cheryl S Rosenfeld
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA.,Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA.,Data Science and Informatics Institute, University of Missouri, Columbia; MO 65211, USA.,Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO, 65211, USA
| |
Collapse
|
4
|
Paul EN, Burns GW, Carpenter TJ, Grey JA, Fazleabas AT, Teixeira JM. Transcriptome Analyses of Myometrium from Fibroid Patients Reveals Phenotypic Differences Compared to Non-Diseased Myometrium. Int J Mol Sci 2021; 22:3618. [PMID: 33807176 PMCID: PMC8036618 DOI: 10.3390/ijms22073618] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/24/2021] [Accepted: 03/28/2021] [Indexed: 12/30/2022] Open
Abstract
Uterine fibroid tissues are often compared to their matched myometrium in an effort to understand their pathophysiology, but it is not clear whether the myometria of uterine fibroid patients represent truly non-disease control tissues. We analyzed the transcriptomes of myometrial samples from non-fibroid patients (M) and compared them with fibroid (F) and matched myometrial (MF) samples to determine whether there is a phenotypic difference between fibroid and non-fibroid myometria. Multidimensional scaling plots revealed that M samples clustered separately from both MF and F samples. A total of 1169 differentially expressed genes (DEGs) (false discovery rate < 0.05) were observed in the MF comparison with M. Overrepresented Gene Ontology terms showed a high concordance of upregulated gene sets in MF compared to M, particularly extracellular matrix and structure organization. Gene set enrichment analyses showed that the leading-edge genes from the TGFβ signaling and inflammatory response gene sets were significantly enriched in MF. Overall comparison of the three tissues by three-dimensional principal component analyses showed that M, MF, and F samples clustered separately from each other and that a total of 732 DEGs from F vs. M were not found in the F vs. MF, which are likely understudied in the pathogenesis of uterine fibroids and could be key genes for future investigation. These results suggest that the transcriptome of fibroid-associated myometrium is different from that of non-diseased myometrium and that fibroid studies should consider using both matched myometrium and non-diseased myometrium as controls.
Collapse
Affiliation(s)
| | | | | | | | | | - Jose M. Teixeira
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University College of Human Medicine, Grand Rapids, MI 49503, USA; (E.N.P.); (G.W.B.); (T.J.C.); (J.A.G.); (A.T.F.)
| |
Collapse
|
5
|
FANCA Polymorphism Is Associated with the Rate of Proliferation in Uterine Leiomyoma in Korea. J Pers Med 2020; 10:jpm10040228. [PMID: 33202820 PMCID: PMC7712130 DOI: 10.3390/jpm10040228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/27/2022] Open
Abstract
Uterine leiomyomas are the most common benign gynecologic tumors. This study was aimed to identify single nucleotide polymorphism of Fanconi anemia complementation group A (FANCA), associated with the rate of proliferation in uterine leiomyomas. In vitro study of patient-derived primary-cultured leiomyoma cells and direct sequencing of fresh frozen leiomyoma from each subject was conducted. Leiomyomas obtained from 44 patients who had underwent surgery were both primary-cultured and freshly frozen. Primary-cultured leiomyoma cells were divided into, according to the rate of proliferation, fast and slow groups. Single nucleotide polymorphism (SNP) of FANCA were determined from fresh frozen tissues of each patient using direct sequencing. Direct sequencing revealed a yet unidentified role of FANCA, a caretaker in the DNA damage-response pathway, as a possible biomarker molecule for the prediction of uterine leiomyoma proliferation. We identified that rs2239359 polymorphism, which causes a missense mutation in FANCA, is associated with the rate of proliferation in uterine leiomyomas. The frequency of C allele in the fast group was 35.29% while that in slow group was 11.11% (odds ratio (OR) 4.036 (1.176–13.855), p = 0.0266). We also found that the TC + CC genotype was more frequently observed in the fast group compared with that in the slow group (OR 6.44 (1.90–31.96), p = 0.0227). Taken together, the results in the current study suggested that a FANCA missense mutation may play an important regulatory role in the proliferation of uterine leiomyoma and thus may serve as a prognostic marker.
Collapse
|
6
|
Mello JBH, Barros-Filho MC, Abreu FB, Cirilo PDR, Domingues MAC, Pontes A, Rogatto SR. MicroRNAs involved in the HMGA2 deregulation and its co-occurrence with MED12 mutation in uterine leiomyoma. Mol Hum Reprod 2019; 24:556-563. [PMID: 30376129 DOI: 10.1093/molehr/gay037] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/21/2018] [Indexed: 12/13/2022] Open
Abstract
STUDY QUESTION Can the mediator complex subunit 12 (MED12) mutation and high mobility group AT-hook 2 (HMGA2) overexpression co-occurrence be explained by the alternative mechanism of HMGA2 dysregulation in uterine leiomyomas (UL)? SUMMARY ANSWER The co-occurrence of MED12 mutation and HMGA2 overexpression, and a negative correlation of five validated or predicted microRNAs that target HMGA2 were reported. WHAT IS KNOWN ALREADY The recent stratification of UL, according to recurrent and mutually exclusive genomic alterations affecting HMGA2, MED12, fumarate hydratase (FH) and collagen type IV alpha 5-alpha 6 (COL4A5-COL4A6) pointed out the involvement of distinct molecular pathways. However, the mechanisms of regulation involving these drivers are poorly explored. STUDY DESIGN, SIZE, DURATION A total of 78 UL and 34 adjacent normal myometrium (NM) tissues was collected from 56 patients who underwent hysterectomies at a single institution. The patients were treated at the Department of Gynecology and Obstetrics, School of Medicine, Sao Paulo State University, Botucatu, SP, Brazil, from October 1995 to February 2004. PARTICIPANTS/MATERIALS, SETTING, METHODS Gene expression profiling was evaluated from fresh frozen tissues and compared with MED12 mutations at exon 2. In addition, RT-qPCR was applied to evaluate the expression levels of HMGA2 and their predictive miRNA regulators: hsa-let-7a, miR-26a, miR-26b, mir-93 and mir-106b. MAIN RESULTS AND THE ROLE OF CHANCE An unsupervised hierarchical clustering analysis revealed two main clusters with one of them (26 of 42 UL) showing an enrichment of MED12 mutated cases (18 of 26 UL). Increased expression levels of HMGA2 were observed in both clusters, including cases with MED12 mutation (cluster 1:18 UL). A significant HMGA2 overexpression (P < 0.001) in UL in comparison with NM was found. Five miRNAs predicted to regulate HMGA2 were significantly downregulated (P < 0.001) and negatively correlated to HMGA2 expression levels (P < 0.05) in UL. LIMITATIONS REASONS FOR CAUTION An in vivo functional study was not performed to validate the microRNAs and HMGA2 interaction due to technical limitations. WIDER IMPLICATIONS OF THE FINDINGS HMGA2 overexpression was detected in a significant number of MED12 mutated ULs, suggesting that these alterations coexist. Furthermore, five miRNAs were described as potential regulators of HMGA2 expression in UL. LARGE-SCALE DATA Data available in the Gene Expression Omnibus GSE42939. STUDY FUNDING AND COMPETING INTEREST(S) This study was supported by grants from Fundação de Amparo a Pesquisa do Estado de São Paulo (# 2008/58835-2) and Conselho Nacional de Pesquisa (# 485032/2007-4), Brazil. The authors declared having no conflicts of interest.
Collapse
Affiliation(s)
- J B H Mello
- CIPE-International Research Center-AC Camargo Cancer Center, Sao Paulo, SP, Brazil
| | - M C Barros-Filho
- CIPE-International Research Center-AC Camargo Cancer Center, Sao Paulo, SP, Brazil
| | - F B Abreu
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - P D R Cirilo
- Hermes Pardini Institute, Research & Development Department, Belo Horizonte, MG, Brazil
| | - M A C Domingues
- Department of Pathology, School of Medicine, University of Sao Paulo State-UNESP, Botucatu-SP, Brazil
| | - A Pontes
- Department of Gynecology and Obstetrics, School of Medicine, University of Sao Paulo State-UNESP, Botucatu-SP, Brazil
| | - S R Rogatto
- Department of Clinical Genetics, Vejle Hospital, Institute of Regional Health Research, University of Southern Denmark, DK, Denmark
| |
Collapse
|
7
|
Lapa RML, Barros-Filho MC, Marchi FA, Domingues MAC, de Carvalho GB, Drigo SA, Kowalski LP, Rogatto SR. Integrated miRNA and mRNA expression analysis uncovers drug targets in laryngeal squamous cell carcinoma patients. Oral Oncol 2019; 93:76-84. [PMID: 31109699 DOI: 10.1016/j.oraloncology.2019.04.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/20/2019] [Accepted: 04/24/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVES The current treatment of laryngeal squamous cell carcinoma (LSCC) is based on radical surgery and radiotherapy resulting in high morbidity. Chemoradiotherapy has been used as alternative to organ sparing; however, several advanced cases presented resistance to treatment, which contributes to a high risk of recurrence and mortality. Coding RNAs and miRNAs have potential to be used as biomarkers or targets for cancer therapy. MATERIALS AND METHODS In this study, 36 LSCC and 5 non-neoplastic control samples were investigated using miRNA and mRNA large-scale expression analysis and a cross-validation was performed using the TCGA database (116 LSCC and 12 surrounding normal tissues). RESULTS The large-scale profiling revealed the involvement of 28 miRNAs and 817 genes differentially expressed in LSCC. An integrative analysis comprising predicted and experimentally validated miRNA/mRNA interactions (negatively correlated), resulted in 28 miRNAs and 543 mRNAs. Decreased expression of miR-199b was significantly associated with shorter disease-free survival in LSCC (internal and TCGA datasets). The expression levels of selected miRNAs (miR-199b-5p, miR-29c-3p, miR-204-5p, miR-125b-5p and miR-92a-3p) and genes (COL3A1, COL10A1, ERBB4, HMGA2, HLF, TOP2A, MMP3, MMP13, MMP10 and PPP1R3) were confirmed as altered in LSCC by RT-qPCR. Additionally, a drug target prediction analysis revealed drug combinations based on miRNA and mRNA expression, pointing out novel alternatives to optimize the LSCC treatment. CONCLUSION Collectively, these findings provide new insights in the LSCC transcriptional deregulation and potential drug targets.
Collapse
Affiliation(s)
- Rainer Marco Lopez Lapa
- International Research Center, CIPE - A.C.Camargo Cancer Center, São Paulo, Brazil; Department of Genetics, Institute of Bioscience, São Paulo State University - UNESP, Botucatu, SP, Brazil
| | | | | | | | | | - Sandra Aparecida Drigo
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University - UNESP, Botucatu, SP, Brazil
| | - Luiz Paulo Kowalski
- Department of Head and Neck Surgery and Otorhinolaryngology, A.C.Camargo Cancer Center, São Paulo Brazil
| | - Silvia Regina Rogatto
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University - UNESP, Botucatu, SP, Brazil; Department of Clinical Genetics, Vejle Hospital, Institute of Regional Health Research, University of Southern Denmark, Denmark.
| |
Collapse
|
8
|
Barros-Filho M, Reis-Rosa L, Hatakeyama M, Marchi F, Chulam T, Scapulatempo-Neto C, Nicolau U, Carvalho A, Pinto C, Drigo S, Kowalski L, Rogatto S. Oncogenic drivers in 11q13 associated with prognosis and response to therapy in advanced oropharyngeal carcinomas. Oral Oncol 2018; 83:81-90. [DOI: 10.1016/j.oraloncology.2018.06.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/26/2018] [Accepted: 06/12/2018] [Indexed: 12/13/2022]
|
9
|
Kuasne H, Barros-Filho MC, Busso-Lopes A, Marchi FA, Pinheiro M, Muñoz JJM, Scapulatempo-Neto C, Faria EF, Guimarães GC, Lopes A, Trindade-Filho JCS, Domingues MAC, Drigo SA, Rogatto SR. Integrative miRNA and mRNA analysis in penile carcinomas reveals markers and pathways with potential clinical impact. Oncotarget 2017; 8:15294-15306. [PMID: 28122331 PMCID: PMC5362487 DOI: 10.18632/oncotarget.14783] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 01/10/2017] [Indexed: 12/14/2022] Open
Abstract
Penile carcinoma (PeCa) is an important public health issue in poor and developing countries, and has only recently been explored in terms of genetic and epigenetic studies. Integrative data analysis is a powerful method for the identification of molecular drivers involved in cancer development and progression. miRNA and mRNA expression profiles followed by integrative analysis were investigated in 23 PeCa and 12 non-neoplastic penile tissues (NPT). Expression levels of eight miRNAs and 10 mRNAs were evaluated in the same set of samples used for microarray and in a validation set of cases (PeCa = 36; NPT = 27). Eighty-one miRNAs and 2,697 mRNAs were identified as differentially expressed in PeCa. Integrative data analysis revealed 255 mRNAs potentially regulated by 68 miRNAs. Using RT-qPCR, eight miRNAs and nine transcripts were confirmed as altered in PeCa. We identified that MMP1, MMP12 and PPARG and hsa-miR-31-5p, hsa-miR-224-5p, and hsa-miR-223-3p were able to distinguish tumors from NPT with high sensitivity and specificity. Higher MMP1 expression was detected as a better predictor of lymph node metastasis than the clinical-pathological data. In addition, PPARG and EGFR were highlighted as potential pathways for targeted therapy in PeCa. The analysis based on HPV positivity (7 of 23 cases) revealed five miRNA and 13 mRNA differentially expressed. Although in a limited number of cases, HPV positive PeCa presented less aggressive phenotype in comparison with negative cases. Overall, an integrative analysis using mRNA and miRNA profiles revealed markers related with tumor development and progression. Furthermore, MMP1 expression level was a predictive marker for lymph node metastasis in patients with PeCa.
Collapse
Affiliation(s)
- Hellen Kuasne
- CIPE-A. C. Camargo Cancer Center, São Paulo, Brazil.,Department of Urology, Faculty of Medicine, São Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| | | | | | | | | | | | | | - Eliney F Faria
- Department of Urology, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | | | - Ademar Lopes
- Department of Urology, A. C. Camargo Cancer Center, São Paulo, Brazil
| | - José C S Trindade-Filho
- Department of Urology, Faculty of Medicine, São Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| | | | - Sandra A Drigo
- Department of Urology, Faculty of Medicine, São Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| | - Silvia R Rogatto
- CIPE-A. C. Camargo Cancer Center, São Paulo, Brazil.,Department of Urology, Faculty of Medicine, São Paulo State University-UNESP, Botucatu, São Paulo, Brazil.,Department of Clinical Genetics, Vejle Sygehus, Vejle, Denmark.,Institute of Regional Health, University of Southern Denmark, Denmark
| |
Collapse
|
10
|
Uterine Inflammatory Myofibroblastic Tumors Frequently Harbor ALK Fusions With IGFBP5 and THBS1. Am J Surg Pathol 2017; 41:773-780. [PMID: 28490045 DOI: 10.1097/pas.0000000000000801] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Inflammatory myofibroblastic tumor (IMT) can occur in a number of anatomic sites, including the uterus. Like its soft tissue counterpart, uterine IMT frequently expresses ALK and harbors ALK genetic rearrangements. The aim of this study is to fully characterize the genetic fusions that occur in uterine IMT. We studied 11 uterine IMTs with typical histology and 8 uterine myxoid smooth muscle tumors (5 leiomyomas, 1 smooth muscle tumor of uncertain malignant potential, and 2 leiomyosarcomas) in which the differential of IMT was considered, using a RNA-sequencing-based fusion assay to detect genetic fusions involving ALK, ROS1, RET, NTRK1/3, and other genes. ALK was expressed in 10 of 11 IMTs and 1 tumor initially categorized as a myxoid leiomyoma (granular cytoplasmic staining with paranuclear accentuation). Fusion transcripts involving ALK were identified in 9 of 10 ALK immunopositive IMTs, with 3 harboring IGFBP5-ALK, 3 harboring THBS1-ALK, 2 harboring FN1-ALK, and 1 harboring TIMP3-ALK. Among the smooth muscle tumors, IGFBP5-ALK fusion transcript was identified in only 1 ALK immunopositive case. Further review revealed that although a diagnosis of IMT was considered for the ALK immunopositive myxoid leiomyoma, this diagnosis was not initially rendered only because fluorescence in situ hybridization analysis was interpreted as negative for ALK genetic rearrangement; this case is best reclassified as an IMT. Notably, all the ALK fusions identified in our study included the transmembrane domain-encoding exon 19 of ALK. Our findings confirm the high frequency of ALK fusions in uterine IMT, with an enrichment of novel 5' ALK fusion partners (IGFBP5, THBS1, and TIMP3) and exon 19-containing ALK fusion. Given that IGFBP5 and FN1 are both situated on the same chromosome as ALK, fluorescence in situ hybridization analysis for ALK rearrangement may not be reliable and a negative result should not exclude a diagnosis of uterine IMT if the histologic features and ALK immunostaining findings are supportive.
Collapse
|
11
|
Lavorato-Rocha AM, Akagi EM, de Melo Maia B, Rodrigues IS, Botelho MCS, Marchi FA, Fernandes G, Baiocchi G, Soares FA, Rogatto SR, Sato-Kuwabara Y, Rocha RM. An Integrative Approach Uncovers Biomarkers that Associate with Clinically Relevant Disease Outcomes in Vulvar Carcinoma. Mol Cancer Res 2016; 14:720-9. [PMID: 27170308 DOI: 10.1158/1541-7786.mcr-15-0366] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 04/21/2016] [Indexed: 11/16/2022]
Abstract
UNLABELLED Vulvar squamous cell carcinoma (VSCC) is a rare disease that has a high mortality rate (∼40%). However, little is known about its molecular signature. Therefore, an integrated genomics approach, based on comparative genome hybridization (aCGH) and genome-wide expression (GWE) array, was performed to identify driver genes in VSCC. To achieve that, DNA and RNA were extracted from frozen VSCC clinical specimens and examined by aCGH and GWE array, respectively. On the basis of the integration of data using the CONEXIC algorithm, PLXDC2 and GNB3 were validated by RT-qPCR. The expression of these genes was then analyzed by IHC in a large set of formalin-fixed paraffin-embedded specimens. These analyses identified 47 putative drivers, 46 of which were characterized by copy number gains that were concomitant with overexpression and one with a copy number loss and downregulation. Two of these genes, PLXDC2 and GNB3, were selected for further validation: PLXDC2 was downregulated and GNB3 was overexpressed compared with non-neoplastic tissue. By IHC, both proteins were ubiquitously expressed throughout vulvar tissue. High expression of GNB3 and low PLXDC2 immunostaining in the same sample was significantly associated with less lymph node metastasis and greater disease-free survival. On the basis of a robust methodology never used before for VSCC evaluation, two novel prognostic markers in vulvar cancer are identified: one with favorable prognosis (GNB3) and the other with unfavorable prognosis (PLXDC2). IMPLICATIONS This genomics study reveals markers that associate with prognosis and may provide guidance for better treatment in vulvar cancer. Mol Cancer Res; 14(8); 720-9. ©2016 AACR.
Collapse
Affiliation(s)
| | - Erica M Akagi
- Molecular Morphology Laboratory, AC Camargo Cancer Center, São Paulo, Brazil
| | | | - Iara S Rodrigues
- Molecular Morphology Laboratory, AC Camargo Cancer Center, São Paulo, Brazil
| | | | - Fabio A Marchi
- NeoGene Laboratory, AC Camargo Cancer Center, São Paulo, Brazil
| | | | - Glauco Baiocchi
- Department of Gynecology Oncology, AC Camargo Cancer Center, São Paulo, Brazil
| | - Fernando A Soares
- Department of Anatomic Pathology, AC Camargo Cancer Center, São Paulo, Brazil
| | | | - Yukie Sato-Kuwabara
- Department of Anatomic Pathology, AC Camargo Cancer Center, São Paulo, Brazil
| | - Rafael M Rocha
- Molecular Morphology Laboratory, AC Camargo Cancer Center, São Paulo, Brazil.
| |
Collapse
|
12
|
Gryshchenko A, Tarnavskiy S, Levchenko K, Bdzhola V, Volynets G, Golub A, Ruban T, Vygranenko K, Lukash L, Yarmoluk S. Design, synthesis and biological evaluation of 5-amino-4-(1H-benzoimidazol-2-yl)-phenyl-1,2-dihydro-pyrrol-3-ones as inhibitors of protein kinase FGFR1. Bioorg Med Chem 2016; 24:2053-9. [DOI: 10.1016/j.bmc.2016.03.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/18/2016] [Accepted: 03/19/2016] [Indexed: 01/27/2023]
|
13
|
Chuang TD, Khorram O. Mechanisms underlying aberrant expression of miR-29c in uterine leiomyoma. Fertil Steril 2016; 105:236-45.e1. [PMID: 26453978 DOI: 10.1016/j.fertnstert.2015.09.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 09/15/2015] [Accepted: 09/15/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To determine the expression of miR-29c and its target genes in leiomyoma and the role of NF-κB, specific protein 1 (SP1), and DNA methylation in its regulation. DESIGN Experimental study. SETTING Academic research laboratory. PATIENT(S) Women undergoing hysterectomy for leiomyoma. INTERVENTION(S) Over- and underexpression of miR-29c; blockade of transcription factors. MAIN OUTCOME MEASURE(S) MiR-29c and its target gene levels in leiomyoma and the effects of blockade of transcription factors on miR-29c expression. RESULT(S) Leiomyoma as compared with myometrium expressed significantly lower levels of miR-29c, with an inverse relationship with expression of its targets, COL3A1 and DNMT3A. Gain of function of miR-29c inhibited the expression of COL3A1 and DNMT3A at protein and mRNA levels, secreted COL3A1, and rate of cell proliferation. Loss of function of miR-29c had the opposite effect. E2, P, and their combination inhibited miR-29c in leiomyoma smooth muscle cells (LSMC). Phosphorylated NF-κB (p65) and SP1 protein expression were significantly increased in leiomyoma. SiRNA knockdown of SP1 and DNMT3A or their specific inhibitors significantly increased the expression of miR-29c, accompanied by the inhibition of cellular and secreted COL3A1 in siRNA-treated cells. Knockdown of p65 also induced miR-29c expression but had no effect on COL3A1 expression. CONCLUSION(S) MiR-29c expression is suppressed in leiomyoma, resulting in an increase in expression of its targets COL3A1 and DNMT3A. The suppression of miR-29c in LSMC is primarily mediated by SP1, NF-κB signaling, and epigenetic modification. Collectively, these results indicate a significant role for miR-29c in leiomyoma pathogenesis.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center and LA-Biomed Research Institute, Torrance, California
| | - Omid Khorram
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center and LA-Biomed Research Institute, Torrance, California.
| |
Collapse
|
14
|
Ling J, Jiang L, Zhang C, Dai J, Wu Q, Tan J. Upregulation of miR-197 inhibits cell proliferation by directly targeting IGFBP5 in human uterine leiomyoma cells. In Vitro Cell Dev Biol Anim 2015; 51:835-42. [PMID: 25990270 DOI: 10.1007/s11626-015-9887-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 03/03/2015] [Indexed: 01/12/2023]
Abstract
Uterine leiomyoma (ULM), one of the most common reproductive tract neoplasms in premenopausal women, is a kind of benign tumor with multigene involved. Finding and studying the key gene involved has been a long-needed factor for developing non-surgery therapy and prevention methods. The dysregulated microRNAs were reported to play important roles in ULM pathobiology by regulating tumor growth. Our investigations have revealed that miR-197 is at low expression in ULM. Characterization of the effects of miR-197 in ULM demonstrated that downregulation of miR-197 increased cell growth and induced cell cycle arrest in the G0/G1 phase in vitro, while upregulation of miR-197 expression had the opposite effect on ULM growth and progression. Further research on the mechanism of miR-197 on the proliferation of ULM cells, we showed that miR-197 inhibited cell proliferation of ULM by directly targeting IGFBP5, which was overexpressed in ULM and played an important role in the etiology of ULM. These findings obtained in this study deliver insights and further expand our understanding of the role of miR-197 and its target IGFBP5 in ULM development, which provides a potential novel therapeutic agent to target the proliferation of ULM cells.
Collapse
Affiliation(s)
- Jing Ling
- Department of Obstetrics and Gynecology, Affiliated Jiangyin Hospital of South-East University, Jiangyin, 214400, China
| | - Li Jiang
- Department of Respiratory, Suzhou Kowloon Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Suzhou, 215021, People's Republic of China
| | - Chenxia Zhang
- Department of Obstetrics and Gynecology, Affiliated Jiangyin Hospital of South-East University, Jiangyin, 214400, China
| | - Jie Dai
- Department of Obstetrics and Gynecology, Affiliated Jiangyin Hospital of South-East University, Jiangyin, 214400, China
| | - Qunying Wu
- Department of Obstetrics and Gynecology, Affiliated Jiangyin Hospital of South-East University, Jiangyin, 214400, China
| | - Jie Tan
- Department of Obstetrics and Gynecology, Affiliated Jiangyin Hospital of South-East University, Jiangyin, 214400, China.
| |
Collapse
|
15
|
Haitjema A, Mol BM, Kooi IE, Massink MPG, Jørgensen JAL, Rockx DAP, Rooimans MA, de Winter JP, Meijers-Heijboer H, Joenje H, Dorsman JC. Coregulation of FANCA and BRCA1 in human cells. SPRINGERPLUS 2014; 3:381. [PMID: 25161863 PMCID: PMC4143540 DOI: 10.1186/2193-1801-3-381] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 06/30/2014] [Indexed: 12/23/2022]
Abstract
Fanconi anemia (FA) is a genetically heterogeneous syndrome associated with increased cancer predisposition. The underlying genes govern the FA pathway which functions to protect the genome during the S-phase of the cell cycle. While upregulation of FA genes has been linked to chemotherapy resistance, little is known about their regulation in response to proliferative stimuli. The purpose of this study was to examine how FA genes are regulated, especially in relation to the cell cycle, in order to reveal their possible participation in biochemical networks. Expression of 14 FA genes was monitored in two human cell-cycle models and in two RB1/E2F pathway-associated primary cancers, retinoblastoma and basal breast cancer. In silico studies were performed to further evaluate coregulation and identify connected networks and diseases. Only FANCA was consistently induced over 2-fold; FANCF failed to exhibit any regulatory fluctuations. Two tools exploiting public data sets indicated coregulation of FANCA with BRCA1. Upregulation of FANCA and BRCA1 correlated with upregulation of E2F3. Genes coregulated with both FANCA and BRCA1 were enriched for MeSH-Term id(s) genomic instability, microcephaly, and Bloom syndrome, and enriched for the cellular component centrosome. The regulation of FA genes appears highly divergent. In RB1-linked tumors, upregulation of FA network genes was associated with reduced expression of FANCF. FANCA and BRCA1 may jointly act in a subnetwork - supporting vital function(s) at the subcellular level (centrosome) as well as at the level of embryonic development (mechanisms controlling head circumference).
Collapse
Affiliation(s)
- Anneke Haitjema
- Department of Clinical Genetics, Section Oncogenetics, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Berber M Mol
- Department of Clinical Genetics, Section Oncogenetics, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Irsan E Kooi
- Department of Clinical Genetics, Section Oncogenetics, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Maarten PG Massink
- Department of Clinical Genetics, Section Oncogenetics, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Jens AL Jørgensen
- Department of Clinical Genetics, Section Oncogenetics, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Davy AP Rockx
- Department of Clinical Genetics, Section Oncogenetics, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Martin A Rooimans
- Department of Clinical Genetics, Section Oncogenetics, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Johan P de Winter
- Department of Clinical Genetics, Section Oncogenetics, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Hanne Meijers-Heijboer
- Department of Clinical Genetics, Section Oncogenetics, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Hans Joenje
- Department of Clinical Genetics, Section Oncogenetics, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Josephine C Dorsman
- Department of Clinical Genetics, Section Oncogenetics, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| |
Collapse
|
16
|
Villacis RAR, Silveira SM, Barros-Filho MC, Marchi FA, Domingues MAC, Scapulatempo-Neto C, Aguiar S, Lopes A, Cunha IW, Rogatto SR. Gene expression profiling in leiomyosarcomas and undifferentiated pleomorphic sarcomas: SRC as a new diagnostic marker. PLoS One 2014; 9:e102281. [PMID: 25028927 PMCID: PMC4100821 DOI: 10.1371/journal.pone.0102281] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 06/17/2014] [Indexed: 12/15/2022] Open
Abstract
Background Undifferentiated Pleomorphic Sarcoma (UPS) and high-grade Leiomyosarcoma (LMS) are soft tissue tumors with an aggressive clinical behavior, frequently developing local recurrence and distant metastases. Despite several gene expression studies involving soft tissue sarcomas, the potential to identify molecular markers has been limited, mostly due to small sample size, in-group heterogeneity and absence of detailed clinical data. Materials and Methods Gene expression profiling was performed for 22 LMS and 22 UPS obtained from untreated patients. To assess the relevance of the gene signature, a meta-analysis was performed using five published studies. Four genes (BAD, MYOCD, SRF and SRC) selected from the gene signature, meta-analysis and functional in silico analysis were further validated by quantitative PCR. In addition, protein-protein interaction analysis was applied to validate the data. SRC protein immunolabeling was assessed in 38 UPS and 52 LMS. Results We identified 587 differentially expressed genes between LMS and UPS, of which 193 corroborated with other studies. Cluster analysis of the data failed to discriminate LMS from UPS, although it did reveal a distinct molecular profile for retroperitoneal LMS, which was characterized by the over-expression of smooth muscle-specific genes. Significantly higher levels of expression for BAD, SRC, SRF, and MYOCD were confirmed in LMS when compared with UPS. SRC was the most value discriminator to distinguish both sarcomas and presented the highest number of interaction in the in silico protein-protein analysis. SRC protein labeling showed high specificity and a positive predictive value therefore making it a candidate for use as a diagnostic marker in LMS. Conclusions Retroperitoneal LMS presented a unique gene signature. SRC is a putative diagnostic marker to differentiate LMS from UPS.
Collapse
Affiliation(s)
- Rolando A. R. Villacis
- Neogene Laboratory, Research Center (CIPE), A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Sara M. Silveira
- Neogene Laboratory, Research Center (CIPE), A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Mateus C. Barros-Filho
- Neogene Laboratory, Research Center (CIPE), A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Fabio A. Marchi
- Inter-Institutional Grad Program on Bioinformatics, Mathematics and Statistics Institute, USP - University of São Paulo, São Paulo, Brazil
| | - Maria A. C. Domingues
- Department of Pathology, School of Medicine, UNESP - São Paulo State University, Botucatu, São Paulo, Brazil
| | | | - Samuel Aguiar
- Department of Pelvic Surgery, A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Ademar Lopes
- Department of Pelvic Surgery, A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Isabela W. Cunha
- Department of Pathology, A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Silvia R. Rogatto
- Neogene Laboratory, Research Center (CIPE), A. C. Camargo Cancer Center, São Paulo, Brazil
- Department of Urology, School of Medicine, UNESP - São Paulo State University, Botucatu, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
17
|
Fujisawa C, Castellot JJ. Matrix production and remodeling as therapeutic targets for uterine leiomyoma. J Cell Commun Signal 2014; 8:179-94. [PMID: 25012731 DOI: 10.1007/s12079-014-0234-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 05/22/2014] [Indexed: 01/06/2023] Open
Abstract
Uterine leiomyoma, commonly known as fibroids, is a benign neoplasm of smooth muscle in women. The incidence of clinically symptomatic fibroids in reproductive-age women is approximately 20 %, with nearly 80 % of black women suffering from this condition. Symptoms include severe pain and hemorrhage; fibroids are also a major cause of infertility or sub-fertility in women. Uterine leiomyoma consist of hyperplastic smooth muscle cells and an excess deposition of extracellular matrix, specifically collagen, fibronectin, and sulfated proteoglycans. Extracellular matrix components interact and signal through integrin-β1 on the surface of uterine leiomyoma smooth muscle cells, provide growth factor storage, and act as co-receptors for growth factor-receptor binding. ECM and growth factor signaling through integrin-β1 and growth factor receptors significantly increases cell proliferation and ECM deposition in uterine leiomyoma. Growth factors TGF-β, IGF, PDGF, FGF and EGF are all shown to promote uterine leiomyoma progression and signal through multiple pathways to increase the expression of genes encoding matrix or matrix-modifying proteins. Decreasing integrin expression, reducing growth factor action and inhibiting ECM action on uterine leiomyoma smooth muscle cells are important opportunities to treat uterine leiomyoma without use of the current surgical procedures. Both natural compounds and chemicals are shown to decrease fibrosis and uterine leiomyoma progression, but further analysis is needed to make inroads in treating this common women's health issue.
Collapse
Affiliation(s)
- Caitlin Fujisawa
- Public Heath and Professional Degrees Program, Tufts University School of Medicine, Boston, MA, 02111, USA
| | | |
Collapse
|
18
|
Karmon AE, Cardozo ER, Rueda BR, Styer AK. MicroRNAs in the development and pathobiology of uterine leiomyomata: does evidence support future strategies for clinical intervention? Hum Reprod Update 2014; 20:670-87. [PMID: 24706045 DOI: 10.1093/humupd/dmu017] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Human leiomyomata (fibroids) are benign tumors of the uterus, represent the most common neoplasms of reproductive-aged women and have a prevalence of ∼70% in the general population. This disorder conveys a significant degree of morbidity and remains the leading indication for hysterectomy in the USA. Prior investigations of aberrant microRNA (miRNA) expression in various malignancies have provided invaluable insight into the role of this class of small non-coding RNAs in tumor growth. Evidence of irregular miRNA expression in uterine fibroids has garnered recent interest for diagnostic and therapeutic applications. Since miRNA gene targets modulate several processes implicated in the genesis of uterine fibroids, more focused investigation has the potential to elucidate the functional significance of miRNA in the genesis and pathology of the disease. METHODS Comprehensive electronic searches of peer reviewed published literature in PubMed (US National Library of Medicine, National Institute of Health; http://www.ncbi.nlm.nih.gov/pubmed/) were performed for content related to the biologic functions of miRNA, the roles of miRNA in human disease and studies investigating miRNA in the context of uterine leiomyomata. Herein, this article will review the current evidence supporting the use of miRNA expression profiling as an investigative tool to assess the pathobiology of uterine fibroids and will discuss potential future applications of miRNAs as biomarkers and therapeutic targets. RESULTS Mounting evidence supports a functional role for miRNA as either indirect or direct regulators of gene expression which impacts the pathobiology of uterine fibroids. Specifically, miRNAs let-7, 200a, 200c, 93, 106b and 21 have been implicated in cellular proliferation, apoptosis, extracellular matrix turnover, angiogenesis and inflammation. Preliminary data provide evidence to suggest that respective in vitro miRNA expression in leiomyomata and myometrium is regulated by sex steroids. CONCLUSIONS Collectively, the identification of aberrantly expressed miRNAs in uterine leiomyomata and accumulating data derived from mining of gene target prediction models and recent functional studies support the concept that miRNAs might impact the genesis and progression of disease. However, the specific biologic functions of differential miRNA expression have yet to be confirmed in vivo. Further functional studies and developing miRNA technology may provide the basis for future applications of miRNAs in clinical medicine as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Anatte E Karmon
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Eden R Cardozo
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Bo R Rueda
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Aaron K Styer
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
19
|
Strotman L, O'Connell R, Casavant BP, Berry SM, Sperger JM, Lang JM, Beebe DJ. Selective nucleic acid removal via exclusion (SNARE): capturing mRNA and DNA from a single sample. Anal Chem 2013; 85:9764-70. [PMID: 24016179 PMCID: PMC3897163 DOI: 10.1021/ac402162r] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The path from gene (DNA) to gene product (RNA or protein) is the foundation of genotype giving rise to phenotype. Comparison of genomic analyses (DNA) with paired transcriptomic studies (mRNA) is critical to evaluating the pathogenic processes that give rise to human disease. The ability to analyze both DNA and mRNA from the same sample is not only important for biologic interrogation but also to minimize variance (e.g., sample loss) unrelated to the biology. Existing methods for RNA and DNA purification from a single sample are typically time-consuming and labor intensive or require large sample sizes to split for separate RNA and DNA extraction procedures. Thus, there is a need for more efficient and cost-effective methods to purify both RNA and DNA from a single sample. To address this need, we have developed a technique, termed SNARE (Selective Nucleic Acid Removal via Exclusion), that uses pinned oil interfaces to simultaneous purify mRNA and DNA from a single sample. A unique advantage of SNARE is the elimination of dilutive wash and centrifugation processes that are fundamental to conventional methods where sample is typically discarded. This minimizes loss and maximizes recovery by allowing nondilutive reinterrogation of the sample. We demonstrate that SNARE is more sensitive than commercially available kits, robustly and repeatably achieving mRNA and DNA purification from extremely low numbers of cells for downstream analyses. In addition to sensitivity, SNARE is fast, easy to use, and cost-effective and requires no laboratory infrastructure or hazardous chemicals. We demonstrate the clinical utility of the SNARE with prostate cancer circulating tumor cells to demonstrate its ability to perform both genomic and transcriptomic interrogation on rare cell populations that would be difficult to achieve with any current method.
Collapse
Affiliation(s)
- Lindsay Strotman
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison , 1111 Highland Avenue, Madison, Wisconsin53706, United States
| | | | | | | | | | | | | |
Collapse
|