1
|
Khazem F, Zetoune AB. Decoding high mobility group A2 protein expression regulation and implications in human cancers. Discov Oncol 2024; 15:322. [PMID: 39085703 PMCID: PMC11291832 DOI: 10.1007/s12672-024-01202-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/29/2024] [Indexed: 08/02/2024] Open
Abstract
High Mobility Group A2 (HMGA2) oncofetal proteins are a distinct category of Transcription Factors (TFs) known as "architectural factors" due to their lack of direct transcriptional activity. Instead, they modulate the three-dimensional structure of chromatin by binding to AT-rich regions in the minor grooves of DNA through their AT-hooks. This binding allows HMGA2 to interact with other proteins and different regions of DNA, thereby regulating the expression of numerous genes involved in carcinogenesis. Consequently, multiple mechanisms exist to finely control HMGA2 protein expression at various transcriptional levels, ensuring precise concentration adjustments to maintain cellular homeostasis. During embryonic development, HMGA2 protein is highly expressed but becomes absent in adult tissues. However, recent studies have revealed its re-elevation in various cancer types. Extensive research has demonstrated the involvement of HMGA2 protein in carcinogenesis at multiple levels. It intervenes in crucial processes such as cell cycle regulation, apoptosis, angiogenesis, epithelial-to-mesenchymal transition, cancer cell stemness, and DNA damage repair mechanisms, ultimately promoting cancer cell survival. This comprehensive review provides insights into the HMGA2 protein, spanning from the genetic regulation to functional protein behavior. It highlights the significant mechanisms governing HMGA2 gene expression and elucidates the molecular roles of HMGA2 in the carcinogenesis process.
Collapse
Affiliation(s)
- Farah Khazem
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Damascus University, Damascus, Syria.
| | | |
Collapse
|
2
|
Saini P, Holmes AG, Wei JJ, Parker JB, Chakravarti D. Engineered uterine primary myometrial cells with high-mobility group AT-hook 2 overexpression display a leiomyoma-like transcriptional and epigenomic phenotype. F&S SCIENCE 2024:S2666-335X(24)00043-0. [PMID: 39074663 DOI: 10.1016/j.xfss.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
OBJECTIVE To determine if engineered high-mobility group AT-hook 2 (HMGA2) overexpressing uterine primary myometrial cells recapitulate the transcriptional and epigenomic features of HMGA2-subtype leiomyomas. DESIGN Isolated primary, "normal" myometrial cells from three patients were engineered to overexpress HMGA2 to determine how HMGA2 establishes transcriptomic and epigenomic features of HMGA2-overexpressing leiomyoma. SETTING Academic research laboratory. PATIENT(S) Primary myometrial cells were isolated from normal myometrium obtained from three patients undergoing hysterectomy. INTERVENTION(S) Not applicable. MAIN OUTCOME MEASURE(S) Determined genome-wide transcriptomic and epigenomic features of engineered HMGA2-overexpressing uterine primary myometrial cells. RESULT(S) Engineered HMGA2-V5-overexpressing primary myometrial cells approximated the HMGA2 expression level observed in HMGA2-overexpression subtype leiomyoma. High-mobility group AT-hook 2-V5 expression resulted in differential expression of 1,612 genes (false discovery rate [FDR] < 0.05) that were found to be enriched in pathways associated with leiomyoma formation, including extracellular matrix organization. Comparative gene expression analysis between HMGA2-V5 engineered primary cells and HMGA2-overexpression subtype leiomyoma revealed significant overlap of differentially expressed genes. Mechanistically, HMGA2-V5 overexpression resulted in 41,323 regions with differential H3K27ac deposition (FDR < 0.05) and 205,605 regions of altered chromatin accessibility (FDR < 0.05). Transcription factor binding site analysis implicated the AP-1 family of transcription factors. CONCLUSION(S) High-mobility group AT-hook 2 overexpression induces leiomyoma-like transcriptomic and epigenomic modulations in myometrial cells.
Collapse
Affiliation(s)
- Priyanka Saini
- Division of Reproductive Sciences in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Austin G Holmes
- Division of Reproductive Sciences in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jian-Jun Wei
- Division of Reproductive Sciences in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - J Brandon Parker
- Division of Reproductive Sciences in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Debabrata Chakravarti
- Division of Reproductive Sciences in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| |
Collapse
|
3
|
Paul EN, Carpenter TJ, Pavliscak LA, Bennett AZ, Ochoa-Bernal MA, Fazleabas AT, Teixeira JM. HMGA2 overexpression induces plasticity in myometrial cells and a transcriptomic profile more similar to that of uterine fibroids. F&S SCIENCE 2024:S2666-335X(24)00041-7. [PMID: 39025326 DOI: 10.1016/j.xfss.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVE To study the possible role for HMGA2 overexpression in differentiated myometrial cells and its potential to induce a stem cell-like or dedifferentiating phenotype and drive fibroid development. DESIGN Myometrial cells were immortalized and transduced with an HMGA2 lentivirus to produce HMGA2hi cells. In vitro stem cell assays were conducted, and ribonucleic acid from HMGA2hi and control cells as well as fibroid-free myometrial and HMGA2 fibroid (HMGA2F) tissues were submitted for ribonucleic acid sequencing. SETTING University research laboratory. PATIENT(S) Women who underwent hysterectomy for symptomatic uterine fibroids or other gynecological conditions. INTERVENTION(S) Not applicable. MAIN OUTCOME MEASURE(S) In vitro stem cell-like properties from myometrial cell lines. Ribonucleic acid sequencing and collagen production of HMGA2-overexpressing primary leiomyoma tissue and cell lines. RESULT(S) HMGA2hi cells had enhanced self-renewal capacity, decreased proliferation, and a greater ability to differentiate into other mesenchymal cell types. HMGA2hi cells exhibited a stem cell-like signature and shared transcriptomic similarities with HMGA2F. Moreover, dysregulated extracellular matrix pathways were observed in both HMGA2hi cells and HMGA2F. CONCLUSION(S) Our findings show that HMGA2 overexpression may drive myometrial cells to dedifferentiate into a more plastic phenotype and provide evidence for an alternative mechanism for fibroid etiology, suggesting that fibroids arise not only from a mutated stem cell but also from a mutated differentiated myometrial cell.
Collapse
Affiliation(s)
- Emmanuel N Paul
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, Michigan
| | - Tyler J Carpenter
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, Michigan
| | - Laura A Pavliscak
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, Michigan
| | - Abigail Z Bennett
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, Michigan
| | - Maria Ariadna Ochoa-Bernal
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, Michigan
| | - Asgerally T Fazleabas
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, Michigan
| | - Jose M Teixeira
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, Michigan.
| |
Collapse
|
4
|
George JW, Cancino RA, Griffin Miller JL, Qiu F, Lin Q, Rowley MJ, Chennathukuzhi VM, Davis JS. Characterization of m6A Modifiers and RNA Modifications in Uterine Fibroids. Endocrinology 2024; 165:bqae074. [PMID: 38946397 PMCID: PMC11222979 DOI: 10.1210/endocr/bqae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 06/07/2024] [Accepted: 06/20/2024] [Indexed: 07/02/2024]
Abstract
Uterine leiomyoma or fibroids are prevalent noncancerous tumors of the uterine muscle layer, yet their origin and development remain poorly understood. We analyzed RNA expression profiles of 15 epigenetic mediators in uterine fibroids compared to myometrium using publicly available RNA sequencing (RNA-seq) data. To validate our findings, we performed RT-qPCR on a separate cohort of uterine fibroids targeting these modifiers confirming our RNA-seq data. We then examined protein profiles of key N6-methyladenosine (m6A) modifiers in fibroids and their matched myometrium, showing no significant differences in concordance with our RNA expression profiles. To determine RNA modification abundance, mRNA and small RNA from fibroids and matched myometrium were analyzed by ultra-high performance liquid chromatography-mass spectrometry identifying prevalent m6A and 11 other known modifiers. However, no aberrant expression in fibroids was detected. We then mined a previously published dataset and identified differential expression of m6A modifiers that were specific to fibroid genetic subtype. Our analysis also identified m6A consensus motifs on genes previously identified to be dysregulated in uterine fibroids. Overall, using state-of-the-art mass spectrometry, RNA expression, and protein profiles, we characterized and identified differentially expressed m6A modifiers in relation to driver mutations. Despite the use of several different approaches, we identified limited differential expression of RNA modifiers and associated modifications in uterine fibroids. However, considering the highly heterogenous genomic and cellular nature of fibroids, and the possible contribution of single molecule m6A modifications to fibroid pathology, there is a need for greater in-depth characterization of m6A marks and modifiers in a larger and diverse patient cohort.
Collapse
Affiliation(s)
- Jitu W George
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Rosa A Cancino
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jennifer L Griffin Miller
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Fang Qiu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Qishan Lin
- RNA Epitranscriptomics and Proteomics Resource, Department of Chemistry, University at Albany, Albany, NY 12222, USA
| | - M Jordan Rowley
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Varghese M Chennathukuzhi
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - John S Davis
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
| |
Collapse
|
5
|
Zhao J, Liao J, Fan W, Yu Q, Lei Z. Multimodality imaging applications in the diagnosis of and surgical treatment strategy for intravenous leiomyomatosis: a case description and literature analysis. Quant Imaging Med Surg 2024; 14:4281-4287. [PMID: 38846306 PMCID: PMC11151228 DOI: 10.21037/qims-23-1772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/12/2024] [Indexed: 06/09/2024]
Affiliation(s)
- Jie Zhao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Jia Liao
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenliang Fan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Qun Yu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Ziqiao Lei
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
6
|
Olson SL, Akbar RJ, Gorniak A, Fuhr LI, Borahay MA. Hypoxia in uterine fibroids: role in pathobiology and therapeutic opportunities. OXYGEN (BASEL, SWITZERLAND) 2024; 4:236-252. [PMID: 38957794 PMCID: PMC11218552 DOI: 10.3390/oxygen4020013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Uterine fibroids are the most common tumors in females affecting up to 70% of women world-wide, yet targeted therapeutic options are limited. Oxidative stress has recently surfaced as a key driver of fibroid pathogenesis and provides insights into hypoxia-induced cell transformation, extracellular matrix pathophysiology, hypoxic cell signaling cascades, and uterine biology. Hypoxia drives fibroid tumorigenesis through (1) promoting myometrial stem cell proliferation, (2) causing DNA damage propelling transformation of stem cells to tumor initiating cells, and (3) driving excess extracellular matrix (ECM) production. Common fibroid-associated DNA mutations include MED12 mutations, HMGA2 overexpression, and Fumarate hydratase loss of function. Evidence suggests an interaction between hypoxia signaling and these mutations. Fibroid development and growth are promoted by hypoxia-triggered cell signaling via various pathways including HIF-1, TGFβ, and Wnt/β-catenin. Fibroid-associated hypoxia persists due to antioxidant imbalance, ECM accumulation, and growth beyond adequate vascular supply. Current clinically available fibroid treatments do not take advantage of hypoxia-targeting therapies. Growing pre-clinical and clinical studies identify ROS inhibitors, anti-HIF-1 agents, Wnt/β-catenin inhibition, and TGFβ cascade inhibitors as agents that may reduce fibroid development and growth through targeting hypoxia.
Collapse
Affiliation(s)
- Sydney L. Olson
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | | | - Adrianna Gorniak
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Laura I. Fuhr
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Mostafa A. Borahay
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| |
Collapse
|
7
|
Paul EN, Carpenter TJ, Pavliscak LA, Bennett AZ, Ochoa-Bernal MA, Fazleabas AT, Teixeira JM. Unraveling the Molecular Landscape of Uterine Fibroids, Insights into HMGA2 and Stem Cell Involvement. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.591351. [PMID: 38712187 PMCID: PMC11071509 DOI: 10.1101/2024.04.26.591351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Uterine fibroids are prevalent benign tumors in women that exhibit considerable heterogeneity in clinical presentation and molecular characteristics, necessitating a deeper understanding of their etiology and pathogenesis. HMGA2 overexpression has been associated with fibroid development, yet its precise role remains elusive. Mutations in fibroids are mutually exclusive and largely clonal, suggesting that tumors originate from a single mutant cell. We explored a possible role for HMGA2 overexpression in differentiated myometrial cells, hypothesizing its potential to induce a stem cell-like or dedifferentiating phenotype and drive fibroid development. Myometrial cells were immortalized and transduced with an HMGA2 lentivirus to produce HMGA2hi cells. In vitro stem cell assays were conducted and RNA from HMGA2hi and control cells and fibroid-free myometrial and HMGA2 fibroid (HMGA2F) tissues were submitted for RNA-sequencing. HMGA2hi cells have enhanced self-renewal capacity, decreased proliferation, and have a greater ability to differentiate into other mesenchymal cell types. HMGA2hi cells exhibit a stem cell-like signature and share transcriptomic similarities with HMGA2F. Moreover, dysregulated extracellular matrix pathways are observed in both HMGA2hi cells and HMGA2F. Our findings suggest that HMGA2 overexpression drives myometrial cells to dedifferentiate into a more plastic phenotype and underscore a pivotal role for HMGA2 in fibroid pathogenesis.
Collapse
|
8
|
Inácio Â, Aguiar L, Rodrigues B, Pires P, Ferreira J, Bilhim T, Pisco J, Bicho M, Clara Bicho M. Leiomyoma and the importance of genetic variation on genes related to the vasculature system - CβS, MTHFR, NOS3, CYBA, and ACE1. Eur J Obstet Gynecol Reprod Biol 2024; 294:65-70. [PMID: 38218160 DOI: 10.1016/j.ejogrb.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 12/29/2023] [Accepted: 01/07/2024] [Indexed: 01/15/2024]
Abstract
OBJECTIVE The link between the systemic vasculature system and tumor biology is here investigated by studying the contribution of CβS (844ins68), MTHFR (677C > T), NOS3 (4a/4b), CYBA (C242T), and ACE1 (I/D) genes to leiomyoma onset, uterus and leiomyoma volumes. METHODS DNA samples from 130 women with leiomyomas and 527 from healthy women were genotyped by PCR or PCR-RFLP. Qui-square (χ2) or Fisher's exact test were used to test associations. All the mentioned tests were performed in IBM® SPSS® Statistics Version 28. Statistical significance was defined as a p-value < 0.05. RESULTS Results revealed that CβS (in the codominant and allelic models, p = 0.044 and, p = 0.015, OR = 1.791 [1.114-2.879], respectively), MTHFR (in the codominant, allelic and dominant models, p = 0.009, p = 0.002, OR = 0.585 [0.416-0.824] and p = 0.003, OR = 0.527 [0.346-0.802], respectively) and ACE1 (dominant model, p = 0.045, OR = 0.639 [0.411-0.992]) genes are associated with leiomyoma onset. NOS3 4a4a genotype is associated with a lower uterus volume (p = 0.004). This study also uncovers intriguing epistatic interactions among some genes that further accentuate their roles in disease modulation. Indeed, the epistatic interactions between the CC genotype (MTHFR) and (+/+) (CβS; p = 0.003), 4b4b (NOS3; p = 0.006, OR = 2.050 [1.223-3.439]) or DD (ACE1; p < 0.001, OR = 2.362 [1.438-3.880]) were shown to be associated with the disease, while 4a presence (NOS3) in epistasis with I presence (ACE1), increased the effect protection having just the I allele presence (p = 0.029, OR = 0.446 [0.214-0.930]). CONCLUSIONS We conclude that variation in genes related to the systemic vascular system can play a role in the onset and development of leiomyoma.
Collapse
Affiliation(s)
- Ângela Inácio
- Instituto de Investigação Científica Bento da Rocha Cabral, Calçada Bento Rocha Cabral, 14, 1257-047 Lisboa, Portugal; Laboratório de Genética, Faculdade de Medicina da Universidade de Lisboa, Avenida Egas Moniz, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal; Instituto de Saúde Ambiental, Laboratório Associado TERRA, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal.
| | - Laura Aguiar
- Instituto de Investigação Científica Bento da Rocha Cabral, Calçada Bento Rocha Cabral, 14, 1257-047 Lisboa, Portugal; Laboratório de Genética, Faculdade de Medicina da Universidade de Lisboa, Avenida Egas Moniz, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal; Instituto de Saúde Ambiental, Laboratório Associado TERRA, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Beatriz Rodrigues
- Instituto de Investigação Científica Bento da Rocha Cabral, Calçada Bento Rocha Cabral, 14, 1257-047 Lisboa, Portugal; Laboratório de Genética, Faculdade de Medicina da Universidade de Lisboa, Avenida Egas Moniz, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Patrícia Pires
- Instituto de Investigação Científica Bento da Rocha Cabral, Calçada Bento Rocha Cabral, 14, 1257-047 Lisboa, Portugal; Laboratório de Genética, Faculdade de Medicina da Universidade de Lisboa, Avenida Egas Moniz, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Joana Ferreira
- Instituto de Investigação Científica Bento da Rocha Cabral, Calçada Bento Rocha Cabral, 14, 1257-047 Lisboa, Portugal; Laboratório de Genética, Faculdade de Medicina da Universidade de Lisboa, Avenida Egas Moniz, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal; Instituto de Saúde Ambiental, Laboratório Associado TERRA, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Tiago Bilhim
- Serviço de Radiologia de Intervenção do Hospital Saint Louis, R. Luz Soriano 182, 1200-249 Lisboa, Portugal; Nova Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - João Pisco
- Serviço de Radiologia de Intervenção do Hospital Saint Louis, R. Luz Soriano 182, 1200-249 Lisboa, Portugal
| | - Manuel Bicho
- Instituto de Investigação Científica Bento da Rocha Cabral, Calçada Bento Rocha Cabral, 14, 1257-047 Lisboa, Portugal; Laboratório de Genética, Faculdade de Medicina da Universidade de Lisboa, Avenida Egas Moniz, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal; Instituto de Saúde Ambiental, Laboratório Associado TERRA, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Maria Clara Bicho
- Instituto de Saúde Ambiental, Laboratório Associado TERRA, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal; Instituto de Medicina Preventiva e Saúde Publica, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
9
|
George JW, Cancino RA, Miller JLG, Qiu F, Lin Q, Rowley MJ, Chennathukuzhi VM, Davis JS. Characterization of m 6A modifiers and RNA modifications in uterine fibroids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.07.552278. [PMID: 37609293 PMCID: PMC10441280 DOI: 10.1101/2023.08.07.552278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Uterine leiomyoma or fibroids are the most common prevalent noncancerous tumors of the uterine muscle layer. Common symptoms associated with fibroids include pelvic pain, heavy menstrual bleeding, anemia, and pelvic pressure. These tumors are a leading cause of gynecological care but lack long-term therapy as the origin and development of fibroids are not well understood. Several next-generation sequencing technologies have been performed to identify the underlying genetic and epigenetic basis of fibroids. However, there remains a systemic gap in our understanding of molecular and biological process that define uterine fibroids. Recent epitranscriptomics studies have unraveled RNA modifications that are associated with all forms of RNA and are thought to influence both normal physiological functions and the progression of diseases. We quantified RNA expression profiles by analyzing publicly available RNA-seq data for 15 known epigenetic mediators to identify their expression profile in uterine fibroids compared to myometrium. To validate our findings, we performed RT-qPCR on a separate cohort of uterine fibroids targeting these modifiers confirming our RNA-seq data. We then examined protein profiles of key m6A modifiers in fibroids and their matched myometrium. In concordance with our RNA expression profiles, no significant differences were observed in these proteins in uterine fibroids compared to myometrium. To determine abundance of RNA modifications, mRNA and small RNA from fibroids and matched myometrium were analyzed by UHPLC MS/MS. In addition to the prevalent N6-methyladenosine (m6A), we identified 11 other known modifiers but did not identify any aberrant expression in fibroids. We then mined a previously published dataset and identified differential expression of m6A modifiers that were specific to fibroid genetic sub-type. Our analysis also identified m6A consensus motifs on genes previously identified to be dysregulated in uterine fibroids. Overall, using state-of-the-art mass spectrometry, RNA expression and protein profiles, we characterized and identified differentially expressed m6A modifiers in relation to driver mutations. Despite the use of several different approaches, we identified limited differential expression of RNA modifiers and associated modifications in uterine fibroids. However, considering the highly heterogenous genomic and cellular nature of fibroids, and the possible contribution of single molecule m6A modifications to fibroid pathology, there is a need for greater in-depth characterization of m6A marks and modifiers in a larger and varied patient cohort.
Collapse
Affiliation(s)
- Jitu W. George
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Veterans Affairs Nebraska Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, USA
| | - Rosa A. Cancino
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jennifer L. Griffin Miller
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Fang Qiu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Qishan Lin
- RNA Epitranscriptomics and Proteomics Resource, Department of Chemistry, University at Albany, Albany, NY, United States
| | - M Jordan Rowley
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska
| | - Varghese M. Chennathukuzhi
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - John S. Davis
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Veterans Affairs Nebraska Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, USA
| |
Collapse
|
10
|
Wang Y, Chen Y, Liang J, Jiang M, Zhang T, Wan X, Wu J, Li X, Chen J, Sun J, Hu Y, Huang P, Feng J, Liu T, Sun X. METTL3-mediated m6A modification of HMGA2 mRNA promotes subretinal fibrosis and epithelial-mesenchymal transition. J Mol Cell Biol 2023; 15:mjad005. [PMID: 36945110 PMCID: PMC10603769 DOI: 10.1093/jmcb/mjad005] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 10/01/2022] [Accepted: 11/28/2022] [Indexed: 03/23/2023] Open
Abstract
Subretinal fibrosis is a major cause of the poor visual prognosis for patients with neovascular age-related macular degeneration (nAMD). Myofibroblasts originated from retinal pigment epithelial (RPE) cells through epithelial-mesenchymal transition (EMT) contribute to the fibrosis formation. N6-Methyladenosine (m6A) modification has been implicated in the EMT process and multiple fibrotic diseases. The role of m6A modification in EMT-related subretinal fibrosis has not yet been elucidated. In this study, we found that during subretinal fibrosis in the mouse model of laser-induced choroidal neovascularization, METTL3 was upregulated in RPE cells. Through m6A epitranscriptomic microarray and further verification, high-mobility group AT-hook 2 (HMGA2) was identified as the key downstream target of METTL3, subsequently activating potent EMT-inducing transcription factor SNAIL. Finally, by subretinal injections of adeno-associated virus vectors, we confirmed that METTL3 deficiency in RPE cells could efficiently attenuate subretinal fibrosis in vivo. In conclusion, our present research identified an epigenetic mechanism of METTL3-m6A-HMGA2 in subretinal fibrosis and EMT of RPE cells, providing a novel therapeutic target for subretinal fibrosis secondary to nAMD.
Collapse
Affiliation(s)
- Yuwei Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Yuhong Chen
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Jian Liang
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Mei Jiang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Ting Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Xiaoling Wan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Jiahui Wu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Xiaomeng Li
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Jieqiong Chen
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Junran Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Yifan Hu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Peirong Huang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Jingyang Feng
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai 200080, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| |
Collapse
|
11
|
Peng Y, Guo R, Shi B, Li D. The role of long non-coding RNA H19 in infertility. Cell Death Discov 2023; 9:268. [PMID: 37507391 PMCID: PMC10382492 DOI: 10.1038/s41420-023-01567-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 07/05/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Infertility is defined as the failure to conceive after at least one year of unprotected intercourse. Long non-coding RNAs (lncRNAs) are transcripts that contain more than 200 nucleotides but do not convert into proteins. LncRNAs, particularly lncRNA H19, have been linked to the emergence and progression of various diseases. This review focuses on the role of H19 in infertility caused by polycystic ovary syndrome, endometriosis, uterine fibroids, diminished ovarian reserve, male factor, and assisted reproductive technology-related pathology, highlighting the potential of H19 as a molecular target for the future treatment of infertility.
Collapse
Affiliation(s)
- Yuanyuan Peng
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, 110004, China
| | - Renhao Guo
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Bei Shi
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, 110004, China.
- Department of Physiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
| | - Da Li
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, 110004, China.
| |
Collapse
|
12
|
Kolterud Å, Välimäki N, Kuisma H, Patomo J, Ilves ST, Mäkinen N, Kaukomaa J, Palin K, Kaasinen E, Karhu A, Pasanen A, Bützow R, Heikinheimo O, Kopp Kallner H, Aaltonen LA. Molecular subclass of uterine fibroids predicts tumor shrinkage in response to ulipristal acetate. Hum Mol Genet 2023; 32:1063-1071. [PMID: 36048862 PMCID: PMC10026225 DOI: 10.1093/hmg/ddac217] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/05/2022] [Accepted: 08/28/2022] [Indexed: 11/12/2022] Open
Abstract
Precision medicine carries great potential for management of all tumor types. The aim of this retrospective study was to investigate if the two most common genetically distinct uterine fibroid subclasses, driven by aberrations in MED12 and HMGA2 genes, respectively, influence response to treatment with the progesterone receptor modulator ulipristal acetate. Changes in diameter and mutation status were derived for 101 uterine fibroids surgically removed after ulipristal acetate treatment. A significant difference in treatment response between the two major subclasses was detected. MED12 mutant fibroids had 4.4 times higher odds of shrinking in response to ulipristal acetate treatment as compared to HMGA2 driven fibroids (95% confidence interval 1.37-13.9; P = 0.013), and in a multivariate analysis molecular subclassification was an independent predictive factor. Compatible with this finding, gene expression and DNA methylation analyses revealed subclass specific differences in progesterone receptor signaling. The work provides a proof-of-principle that uterine fibroid treatment response is influenced by molecular subclass and that the genetic subclasses should be taken into account when evaluating current and future uterine fibroid therapies.
Collapse
Affiliation(s)
- Åsa Kolterud
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Niko Välimäki
- Department of Medical and Clinical Genetics, University of Helsinki; Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, 00290 Helsinki, Finland
| | - Heli Kuisma
- Department of Medical and Clinical Genetics, University of Helsinki; Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, 00290 Helsinki, Finland
| | - Joonatan Patomo
- Department of Medical and Clinical Genetics, University of Helsinki; Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, 00290 Helsinki, Finland
| | - Sini T Ilves
- Department of Medical and Clinical Genetics, University of Helsinki; Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, 00290 Helsinki, Finland
| | - Netta Mäkinen
- Department of Medical and Clinical Genetics, University of Helsinki; Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, 00290 Helsinki, Finland
| | - Jaana Kaukomaa
- Department of Medical and Clinical Genetics, University of Helsinki; Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, 00290 Helsinki, Finland
| | - Kimmo Palin
- Department of Medical and Clinical Genetics, University of Helsinki; Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, 00290 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00290 Helsinki, Finland
| | - Eevi Kaasinen
- Department of Medical and Clinical Genetics, University of Helsinki; Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, 00290 Helsinki, Finland
| | - Auli Karhu
- Department of Medical and Clinical Genetics, University of Helsinki; Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, 00290 Helsinki, Finland
| | - Annukka Pasanen
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, 00290 Helsinki, Finland
| | - Ralf Bützow
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, 00290 Helsinki, Finland
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, 00029 HUS Helsinki, Finland
| | - Oskari Heikinheimo
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, 00029 HUS Helsinki, Finland
| | - Helena Kopp Kallner
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Obstetrics and Gynecology, Danderyd Hospital, 182 88 Stockholm, Sweden
| | - Lauri A Aaltonen
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83 Huddinge, Sweden
- Department of Medical and Clinical Genetics, University of Helsinki; Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, 00290 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00290 Helsinki, Finland
| |
Collapse
|
13
|
A View on Uterine Leiomyoma Genesis through the Prism of Genetic, Epigenetic and Cellular Heterogeneity. Int J Mol Sci 2023; 24:ijms24065752. [PMID: 36982825 PMCID: PMC10056617 DOI: 10.3390/ijms24065752] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/07/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Uterine leiomyomas (ULs), frequent benign tumours of the female reproductive tract, are associated with a range of symptoms and significant morbidity. Despite extensive research, there is no consensus on essential points of UL initiation and development. The main reason for this is a pronounced inter- and intratumoral heterogeneity resulting from diverse and complicated mechanisms underlying UL pathobiology. In this review, we comprehensively analyse risk and protective factors for UL development, UL cellular composition, hormonal and paracrine signalling, epigenetic regulation and genetic abnormalities. We conclude the need to carefully update the concept of UL genesis in light of the current data. Staying within the framework of the existing hypotheses, we introduce a possible timeline for UL development and the associated key events—from potential prerequisites to the beginning of UL formation and the onset of driver and passenger changes.
Collapse
|
14
|
Toledano-Fonseca M, Gómez-España MA, Élez E, Grávalos C, García-Alfonso P, Rodríguez R, Losa F, Alés Díaz I, Graña B, Valladares-Ayerbes M, García-Ortiz MV, Polo E, Salgado M, Rivera F, Safont MJ, Salud A, Ruiz-Casado A, Tabernero JM, Riesco MC, Rodríguez-Ariza A, Aranda E. A signature of circulating microRNAs predicts the response to treatment with FOLFIRI plus aflibercept in metastatic colorectal cancer patients. Biomed Pharmacother 2023; 159:114272. [PMID: 36706629 DOI: 10.1016/j.biopha.2023.114272] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
The benefit of adding the antiangiogenic drug aflibercept to FOLFIRI regime in metastatic colorectal cancer (CRC) patients resistant to or progressive on an oxaliplatin-based therapy has been previously demonstrated. However, the absence of validated biomarkers to predict greater outcomes is a major challenge encountered when using antiangiogenic therapies. In this study we investigated profiles of circulating microRNAs (miRNAs) to build predictive models of response to treatment and survival. Plasma was obtained from 98 metastatic CRC patients enrolled in a clinical phase II trial before receiving FOLFIRI plus aflibercept treatment, and the circulating levels of 754 individual miRNAs were quantified using real-time PCR. A distinct signature of circulating miRNAs differentiated responder from non-responder patients. Remarkably, most of these miRNAs were found to target genes that are involved in angiogenic processes. Accordingly, some of these miRNAs had predictive value and entered in predictive models of response to therapy, progression of disease, and survival of patients treated with FOLFIRI plus aflibercept. Among these miRNAs, circulating levels of hsa-miR-33b-5p efficiently discriminated between responder and non-responder patients and predicted the risk of disease progression. Moreover, the combination of circulating VEGF-A and miR-33b-5p levels improved clinical stratification of metastatic CRC patients who were to receive FOLFIRI plus aflibercept treatment. In conclusion, our study supports circulating miRNAs as valuable biomarkers for predicting better outcomes in metastatic CRC patients treated with FOLFIRI plus aflibercept.
Collapse
Affiliation(s)
- M Toledano-Fonseca
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), E14004 Córdoba, Spain; Cancer Network Biomedical Research Center (CIBERONC), Instituto de Salud Carlos III, E28029 Madrid, Spain.
| | - M A Gómez-España
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), E14004 Córdoba, Spain; Department of Medical Oncology, Reina Sofía University Hospital, E14004 Córdoba, Spain.
| | - E Élez
- Department of Medical Oncology, Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119, 08035 Barcelona, Spain.
| | - C Grávalos
- Department of Medical Oncology, H. Universitario 12 de Octubre, Instituto de Investigación i+12, Avenida de Córdoba, S/N, 28041 Madrid, Spain.
| | - P García-Alfonso
- Department of Medical Oncology, H. Gregorio Marañón, Calle del Doctor Esquerdo, 46, 28007 Madrid, Spain.
| | - R Rodríguez
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), E14004 Córdoba, Spain; Department of Medical Oncology, Reina Sofía University Hospital, E14004 Córdoba, Spain.
| | - F Losa
- Department of Medical Oncology, ICO-CSI. H. Sant Joan Despí - Moisés Broggi, Carrer d'Oriol Martorell, 12, 08970 Sant Joan Despí, Barcelona, Spain.
| | - I Alés Díaz
- Department of Medical Oncology, Unidad de Gestión Clínica Intercentros de Oncología Médica. Hospitales Universitarios Regional y Virgen de la Victoria. IBIMA, Campus de Teatinos, S/N, 29010 Málaga, Spain.
| | - B Graña
- Department of Medical Oncology, C. H. Universitario, Lugar, Xubias de Arriba, 84, 15006 A Coruña, Spain.
| | - M Valladares-Ayerbes
- Department of Medical Oncology, H. Virgen del Rocío, IBIS, Av. Manuel Siurot s/n, 41013 Sevilla, Spain.
| | - M V García-Ortiz
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), E14004 Córdoba, Spain; Cancer Network Biomedical Research Center (CIBERONC), Instituto de Salud Carlos III, E28029 Madrid, Spain.
| | - E Polo
- Department of Medical Oncology, H. Miguel Servet, Paseo Isabel la Católica, 1-3, 50009 Zaragoza, Spain.
| | - M Salgado
- Department of Medical Oncology, C. H. Universitario de Ourense, Calle Ramón Puga Noguerol, 54, 32005 Orense, Spain.
| | - F Rivera
- Department of Medical Oncology, H. Universitario Marqués de Valdecilla, IDIVAL, Avenida de Valdecilla, 25, 39008 Santander, Spain.
| | - M J Safont
- Department of Medical Oncology, H. General Universitario, CIBERONC, Universidad de Valencia, Avenida de les Tres Creus, 2, 46014 Valencia, Spain.
| | - A Salud
- Department of Medical Oncology, H. Universitario Arnau de Vilanova, Avenida Alcalde Rovira Roure, 80, 25198 Lérida, Spain.
| | - A Ruiz-Casado
- Department of Medical Oncology, H. Puerta de Hierro Majadahonda, Calle Joaquín Rodrigo, 1, 28222 Majadahonda, Spain.
| | - J M Tabernero
- Department of Medical Oncology, Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119, 08035 Barcelona, Spain.
| | - M C Riesco
- Department of Medical Oncology, H. Universitario 12 de Octubre, Instituto de Investigación i+12, Avenida de Córdoba, S/N, 28041 Madrid, Spain.
| | - A Rodríguez-Ariza
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), E14004 Córdoba, Spain; Cancer Network Biomedical Research Center (CIBERONC), Instituto de Salud Carlos III, E28029 Madrid, Spain; Department of Medical Oncology, Reina Sofía University Hospital, E14004 Córdoba, Spain.
| | - E Aranda
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), E14004 Córdoba, Spain; Cancer Network Biomedical Research Center (CIBERONC), Instituto de Salud Carlos III, E28029 Madrid, Spain; Department of Medical Oncology, Reina Sofía University Hospital, E14004 Córdoba, Spain; Department of Medicine, Faculty of Medicine, University of Córdoba, E14004 Córdoba, Spain.
| | | |
Collapse
|
15
|
Zhang B, You T, Liu Y, Li P. CIRC_0114428 INFLUENCES THE PROGRESSION OF SEPTIC ACUTE KIDNEY INJURY VIA REGULATING MIR-370-3P/TIMP2 AXIS. Shock 2023; 59:505-513. [PMID: 36597773 DOI: 10.1097/shk.0000000000002077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
ABSTRACT Background: Septic acute kidney injury (AKI) is a serious complication of sepsis, which greatly threatened the life safety of critically ill patients. Recently, circular RNA is considered to be implicated in sepsis-induced renal cell damage. However, the role of circ_0114428 in sepsis AKI is still unclear. Methods: LPS was used to establish a sepsis-related AKI cell model. The expression of circ_0114428, microRNA (miR)-370-3p, tissue inhibitor of metalloproteinase-2 (TIMP2), Proliferating cell nuclear antigen, Bax, and Bcl-2 were detected by quantitative real-time polymerase chain reaction and Western blot. Cell counting kit 8 and enzyme-linked immunosorbent assay were used to measure cell proliferation ability and the secretion of inflammatory factors (tumor necrosis factor α, interleukin 1β, and interleukin 6), respectively. Cell cycle and apoptosis rate were analyzed by flow cytometry. Caspase-3 assay kits were used to detect Caspase-3 activity. Interaction between miR-370-3p and circ_0114428 or TIMP2 was analyzed by bioinformatics analysis, a dual-luciferase reporter assay, and RNA immunoprecipitation assay. Results: Circ_0114428 was upregulated in septic AKI serum samples and LPS-induced HK2 cells. The knockdown of circ_0114428 notably promoted cell proliferation and cycle, whereas it restrained cell inflammation and apoptosis in LPS-stimulated HK2 cells. Subsequent mechanism analysis revealed that miR-370-3p was a target of circ_0114428, and miR-370-3p inhibition could rescue the effects of circ_0114428 downregulation on LPS-induced cell injury. Meanwhile, TIMP2 was a target gene of miR-370-3p. miR-370-3p mimic could attenuate LPS-induced cell injury, whereas these impacts were overturned by overexpressed TIMP2. Furthermore, circ_0114428 enhanced TIMP2 protein expression by sponging miR-370-3p. Conclusion: Our data demonstrated that circ_0114428 contributed to septic AKI progression by regulating miR-370-3p-mediated TIMP2 expression, which provided a promising target for septic AKI treatment.
Collapse
Affiliation(s)
- Bihong Zhang
- Department of Emergency, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Ting You
- Department of Emergency, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yu Liu
- Department of Emergency, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Pei Li
- Department of Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
16
|
Ren R, Ma K, Jiang Y, Chen J, Kou Y, Ge Z, Chen Z, Wei X, Yu L. Endothelial miR-196b-5p regulates angiogenesis via the hypoxia/miR-196b-5p/HMGA2/HIF1α loop. Am J Physiol Cell Physiol 2023; 324:C407-C419. [PMID: 36534502 DOI: 10.1152/ajpcell.00309.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Angiogenesis is involved in development, reproduction, wound healing, homeostasis, and other pathophysiological events. Imbalanced angiogenesis predisposes patients to various pathological processes, such as angiocardiopathy, inflammation, and tumorigenesis. MicroRNAs (miRNAs) have been found to be important in regulating cellular processing and physiological events including angiogenesis. However, the role of miRNAs that regulate angiogenesis (angiomiRs) is not fully understood. Here, we observed a downregulation of the miR-196 family in endothelial cells upon hypoxia. Functionally, miR-196b-5p inhibited the angiogenic functions of endothelial cells in vitro and suppressed angiogenesis in Matrigel plugs and skin wound healing in vivo. Mechanistically, miR-196b-5p bound onto the 3' untranslated region (UTR) of high-mobility group AT-hook 2 (HMGA2) mRNA and repressed the translation of HMGA2, which in turn represses HIF1α accumulation in endothelial cells upon hypoxia. Together, our results establish the role of endothelial miR-196b-5p as an angiomiR that negatively regulates endothelial growth in angiogenesis via the hypoxia/miR-196b-5p/HMGA2/HIF1α loop. miR-196b-5p and its regulatory loop could be an important addition to the molecular mechanisms underlying angiogenesis and may serve as potential targets for antiangiogenic therapy.
Collapse
Affiliation(s)
- Ruizhe Ren
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, People's Republic of China.,College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Kefan Ma
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, People's Republic of China.,College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Yuanqing Jiang
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, People's Republic of China.,College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Junbo Chen
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, People's Republic of China.,College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Yaohui Kou
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, People's Republic of China.,College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Zhen Ge
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, People's Republic of China
| | - Zhaoming Chen
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Xiyang Wei
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Luyang Yu
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, People's Republic of China.,College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| |
Collapse
|
17
|
Jokinen V, Mehine M, Reinikka S, Khamaiseh S, Ahvenainen T, Äyräväinen A, Härkki P, Bützow R, Pasanen A, Vahteristo P. 3'RNA and whole-genome sequencing of archival uterine leiomyomas reveal a tumor subtype with chromosomal rearrangements affecting either HMGA2, HMGA1, or PLAG1. Genes Chromosomes Cancer 2023; 62:27-38. [PMID: 35822448 PMCID: PMC9804854 DOI: 10.1002/gcc.23088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 01/09/2023] Open
Abstract
Uterine leiomyomas, or fibroids, are very common smooth muscle tumors that arise from the myometrium. They can be divided into distinct molecular subtypes. We have previously shown that 3'RNA-sequencing is highly effective in classifying archival formalin-fixed paraffin-embedded (FFPE) leiomyomas according to the underlying mutation. In this study, we performed 3'RNA-sequencing with 111 FFPE leiomyomas previously classified as negative for driver alterations in mediator complex subunit 12 (MED12), high mobility group AT-hook 2 (HMGA2), and fumarate hydratase (FH) by Sanger sequencing and immunohistochemistry. This revealed 43 tumors that displayed expression features typically seen in HMGA2-positive tumors, including overexpression of PLAG1. We explored 12 such leiomyomas by whole-genome sequencing to identify their underlying genomic drivers and to evaluate the feasibility of detecting chromosomal driver alterations from FFPE material. Four tumors with significant HMGA2 overexpression at the protein-level served as controls. We identified chromosomal rearrangements targeting either HMGA2, HMGA1, or PLAG1 in all 16 tumors, demonstrating that it is possible to detect chromosomal driver alterations in archival leiomyoma specimens as old as 18 years. Furthermore, two tumors displayed biallelic loss of DEPDC5 and one tumor harbored a COL4A5-COL4A6 deletion. These observations suggest that instead of only HMGA2-positive leiomyomas, a distinct leiomyoma subtype is characterized by rearrangements targeting either HMGA2, HMGA1, or PLAG1. The results indicate that the frequency of HMGA2-positive leiomyomas may be higher than estimated in previous studies where immunohistochemistry has been used. This study also demonstrates the feasibility of detecting chromosomal driver alterations from archival FFPE material.
Collapse
Affiliation(s)
- Vilja Jokinen
- Applied Tumor Genomics Research ProgramUniversity of HelsinkiHelsinkiFinland,Department of Medical and Clinical GeneticsUniversity of HelsinkiHelsinkiFinland
| | - Miika Mehine
- Applied Tumor Genomics Research ProgramUniversity of HelsinkiHelsinkiFinland,Department of Medical and Clinical GeneticsUniversity of HelsinkiHelsinkiFinland
| | - Siiri Reinikka
- Applied Tumor Genomics Research ProgramUniversity of HelsinkiHelsinkiFinland,Department of Medical and Clinical GeneticsUniversity of HelsinkiHelsinkiFinland
| | - Sara Khamaiseh
- Applied Tumor Genomics Research ProgramUniversity of HelsinkiHelsinkiFinland,Department of Medical and Clinical GeneticsUniversity of HelsinkiHelsinkiFinland,iCAN Digital Precision Cancer Medicine FlagshipHelsinkiFinland
| | - Terhi Ahvenainen
- Applied Tumor Genomics Research ProgramUniversity of HelsinkiHelsinkiFinland,Department of Medical and Clinical GeneticsUniversity of HelsinkiHelsinkiFinland,iCAN Digital Precision Cancer Medicine FlagshipHelsinkiFinland
| | - Anna Äyräväinen
- Applied Tumor Genomics Research ProgramUniversity of HelsinkiHelsinkiFinland,Department of Medical and Clinical GeneticsUniversity of HelsinkiHelsinkiFinland,Department of Obstetrics and GynecologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Päivi Härkki
- Department of Obstetrics and GynecologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Ralf Bützow
- Applied Tumor Genomics Research ProgramUniversity of HelsinkiHelsinkiFinland,Department of PathologyUniversity of Helsinki and HUSLAB, Helsinki University HospitalHelsinkiFinland
| | - Annukka Pasanen
- Applied Tumor Genomics Research ProgramUniversity of HelsinkiHelsinkiFinland,Department of PathologyUniversity of Helsinki and HUSLAB, Helsinki University HospitalHelsinkiFinland
| | - Pia Vahteristo
- Applied Tumor Genomics Research ProgramUniversity of HelsinkiHelsinkiFinland,Department of Medical and Clinical GeneticsUniversity of HelsinkiHelsinkiFinland,iCAN Digital Precision Cancer Medicine FlagshipHelsinkiFinland
| |
Collapse
|
18
|
Hu Y, Chen J, Liu M, Feng Q, Peng H. IGF2BP2 serves as a core m6A regulator in head and neck squamous cell carcinoma. Biosci Rep 2022; 42:231970. [PMID: 36281789 PMCID: PMC9653096 DOI: 10.1042/bsr20221311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/07/2022] [Accepted: 10/24/2022] [Indexed: 02/05/2023] Open
Abstract
Methylation of N6 adenosine (m6A) plays a crucial role in the development and progression of cancers. Its modification is regulated by three types of m6A-related regulators (methyltransferases (writers), demethylases (erasers), and RNA-binding proteins (readers)). Till now, the functions and roles of these regulators in head and neck squamous cell carcinoma (HNSC) remain largely unexplored. Therefore, we utilized the open HNSC dataset in The Cancer Genome Atlas (TCGA), four different cell lines, and our HNSC patient samples (n=40) to explore the clinical significance of 19 m6A regulators, and selected the most significant prognosis-related regulator. Authentic analyses based on online websites were also used in the study (Oncomine, UALCAN, Kaplan-Meier plotter, Human Protein Atlas (HPA), cBioPortal, LinkedOmics, String, etc.). From the results, general overexpression of m6A regulators was observed in pan-cancer, especially in HNSC. IGF2BP2 was recognized as the hub m6A regulator, which was an independent, unfavorable prognostic factor in HNSC. Its mRNA and protein expression in HNSC were significantly up-regulated. Gene mutation types of IGF2BP2 in HNSC (32%) were mainly mRNA High or Amplification, which represented the high expression of IGF2BP2. And these mutations were associated with a poor prognosis. In functional analysis, IGF2BP2 was negatively correlated to tumor immune infiltration in HNSC. Finally, HMGA2 might interact with the IGF2BP2 in HNSC. In conclusion, IGF2BP2 serves as a core m6A regulator among all regulators in HNSC, which has a high expression and predicts the poor prognosis of HNSC patients independently. IGF2BP2 might bring a new direction for HNSC treatment in the future.
Collapse
Affiliation(s)
- Yuan Hu
- Department of Head and Neck Surgery, Cancer Hospital of Shantou University Medical College, Guangdong 515041, P.R. China
| | - Jiexin Chen
- Department of Rheumatology, First Affiliated Hospital of Shantou University Medical College, Guangdong 515041, P.R. China
| | - Muyuan Liu
- Department of Head and Neck Surgery, Cancer Hospital of Shantou University Medical College, Guangdong 515041, P.R. China
| | - Qin Feng
- Department of Head and Neck Surgery, Cancer Hospital of Shantou University Medical College, Guangdong 515041, P.R. China
| | - Hanwei Peng
- Department of Head and Neck Surgery, Cancer Hospital of Shantou University Medical College, Guangdong 515041, P.R. China
| |
Collapse
|
19
|
The proteasome activator REGγ promotes diabetic endothelial impairment by inhibiting HMGA2-GLUT1 pathway. Transl Res 2022; 246:33-48. [PMID: 35367424 DOI: 10.1016/j.trsl.2022.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 11/22/2022]
Abstract
Diabetic vascular endothelial impairment is one of the main causes of death in patients with diabetes lacking adequately defined mechanisms or effective treatments. REGγ, the 11S proteasome activator known to promote the degradation of cellular proteins in a ubiquitin- and ATP-independent manner, emerges as a new regulator in the cardiovascular system. Here, we found that REGγ was upregulated in streptozocin (STZ)-induced diabetic mouse aortic endothelium in vivo and high glucose (HG)-treated vascular endothelial cells (ECs) in vitro. REGγ deficiency ameliorated endothelial impairment in STZ-induced diabetic mice by protecting against a decline in cellular glucose uptake and associated vascular ECs dysfunction by suppressing high mobility group AT-hook 2 (HMGA2) decay. Mechanically, REGγ interacted with and degraded the transcription factor HMGA2 directly, leading to decreased HMGA2 transcriptional activity, subsequently lowered expression of glucose transporter type 1 (GLUT1), and reduced cellular glucose uptake, vascular endothelial dysfunction, and impaired diabetic endothelium. Ablation of endogenous GLUT1 or HMGA2 or overexpressing exogenous HMGA2 in vascular ECs significantly blocked or reestablished the REGγ-dependent action on cellular glucose uptake and vascular endothelial functions of HG stimulation in vitro. Furthermore, exogenously introducing HMGA2 improved diabetic mice endothelial impairment features caused by REGγ in vivo, thereby substantiating a REGγ-HMGA2-GLUT1 pathway in diabetic endothelial impairment. Our findings indicate that modulating REGγ-proteasome activity may be a potential therapeutic approach for diabetic disorders with endothelial impairment.
Collapse
|
20
|
Drakopoulou E, Anagnou NP, Pappa KI. Gene Therapy for Malignant and Benign Gynaecological Disorders: A Systematic Review of an Emerging Success Story. Cancers (Basel) 2022; 14:cancers14133238. [PMID: 35805007 PMCID: PMC9265289 DOI: 10.3390/cancers14133238] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/21/2022] [Accepted: 06/27/2022] [Indexed: 12/24/2022] Open
Abstract
Simple Summary This review discusses all the major advances in gene therapy of gynaecological disorders, highlighting the novel and potentially therapeutic perspectives associated with such an approach. It specifically focuses on the gene therapy strategies against major gynaecological malignant disorders, such as ovarian, cervical, and endometrial cancer, as well as benign disorders, such as uterine leiomyomas, endometriosis, placental, and embryo implantation disorders. The above therapeutic strategies, which employ both viral and non-viral systems for mutation compensation, suicide gene therapy, oncolytic virotherapy, antiangiogenesis and immunopotentiation approaches, have yielded promising results over the last decade, setting the grounds for successful clinical trials. Abstract Despite the major advances in screening and therapeutic approaches, gynaecological malignancies still present as a leading cause of death among women of reproductive age. Cervical cancer, although largely preventable through vaccination and regular screening, remains the fourth most common and most lethal cancer type in women, while the available treatment schemes still pose a fertility threat. Ovarian cancer is associated with high morbidity rates, primarily due to lack of symptoms and high relapse rates following treatment, whereas endometrial cancer, although usually curable by surgery, it still represents a therapeutic problem. On the other hand, benign abnormalities, such as fibroids, endometriosis, placental, and embryo implantation disorders, although not life-threatening, significantly affect women’s life and fertility and have high socio-economic impacts. In the last decade, targeted gene therapy approaches toward both malignant and benign gynaecological abnormalities have led to promising results, setting the ground for successful clinical trials. The above therapeutic strategies employ both viral and non-viral systems for mutation compensation, suicide gene therapy, oncolytic virotherapy, antiangiogenesis and immunopotentiation. This review discusses all the major advances in gene therapy of gynaecological disorders and highlights the novel and potentially therapeutic perspectives associated with such an approach.
Collapse
Affiliation(s)
- Ekati Drakopoulou
- Laboratory of Cell and Gene Therapy, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece; (E.D.); (K.I.P.)
| | - Nicholas P. Anagnou
- Laboratory of Cell and Gene Therapy, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece; (E.D.); (K.I.P.)
- Correspondence:
| | - Kalliopi I. Pappa
- Laboratory of Cell and Gene Therapy, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece; (E.D.); (K.I.P.)
- First Department of Obstetrics and Gynecology, University of Athens School of Medicine, 11528 Athens, Greece
| |
Collapse
|
21
|
Salas A, Beltrán-Flores S, Évora C, Reyes R, Montes de Oca F, Delgado A, Almeida TA. Stem Cell Growth and Differentiation in Organ Culture: New Insights for Uterine Fibroid Treatment. Biomedicines 2022; 10:biomedicines10071542. [PMID: 35884847 PMCID: PMC9313456 DOI: 10.3390/biomedicines10071542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 11/18/2022] Open
Abstract
Organ culture allows for the understanding of normal and tumor cell biology, and tissues generally remain viable for 5–7 days. Strikingly, we determined that myometrial and MED12 mutant leiomyoma cells repopulated cell-depleted tissue slices after 20 days of culture. Using immunofluorescence and quantitative PCR of stem cell and undifferentiated cell markers, we observed clusters of CD49b+ cells in tumor slices. CD49b+ cells, however, were sparsely detected in the myometrial slices. Almost all LM cells strongly expressed Ki67, while only a few myometrial cells were stained for this proliferation marker. The CD73 marker was expressed only in tumor cells, whereas the mesenchymal stem cell receptor KIT was detected only in normal cells. HMGA2 and CD24 showed broader expression patterns and higher signal intensity in leiomyoma than in myometrial cells. In this study, we propose that activating CD49b+ stem cells in myometrium leads to asymmetrical division, giving rise to transit-amplifying KIT+ cells that differentiate to smooth muscle cells. On the contrary, activated leiomyoma CD49b+ cells symmetrically divide to form clusters of stem cells that divide and differentiate to smooth muscle cells without losing proliferation ability. In conclusion, normal and mutant stem cells can proliferate and differentiate in long-term organ culture, constituting a helpful platform for novel therapeutic discovery.
Collapse
Affiliation(s)
- Ana Salas
- Department of Biochemistry, Microbiology, Cell Biology and Genetics, Biology Section, Science Faculty, University of La Laguna, Ave. Astrofísico Fco. Sánchez s/n. San Cristóbal de La Laguna, 38200 Santa Cruz de Tenerife, Spain; (A.S.); (S.B.-F.); (R.R.)
- Institute of Tropical Diseases and Healthcare of the Canary Island, Ave. Astrofísico Fco. Sánchez s/n. San Cristóbal de La Laguna, 38200 Santa Cruz de Tenerife, Spain
| | - Silvia Beltrán-Flores
- Department of Biochemistry, Microbiology, Cell Biology and Genetics, Biology Section, Science Faculty, University of La Laguna, Ave. Astrofísico Fco. Sánchez s/n. San Cristóbal de La Laguna, 38200 Santa Cruz de Tenerife, Spain; (A.S.); (S.B.-F.); (R.R.)
| | - Carmen Évora
- Department of Chemical Engineering and Pharmaceutical Technology, Faculty of Pharmacy, University of La Laguna, Ave. Astrofísico Fco. Sánchez s/n. San Cristóbal de La Laguna, 38200 Santa Cruz de Tenerife, Spain; (C.É.); (A.D.)
- Institute of Biomedical Technologies (ITB), Medicine Section, Faculty of Health Science, University of La Laguna, St. Santa María Soledad, s/n. San Cristóbal de La Laguna, 38200 Santa Cruz de Tenerife, Spain
| | - Ricardo Reyes
- Department of Biochemistry, Microbiology, Cell Biology and Genetics, Biology Section, Science Faculty, University of La Laguna, Ave. Astrofísico Fco. Sánchez s/n. San Cristóbal de La Laguna, 38200 Santa Cruz de Tenerife, Spain; (A.S.); (S.B.-F.); (R.R.)
- Institute of Tropical Diseases and Healthcare of the Canary Island, Ave. Astrofísico Fco. Sánchez s/n. San Cristóbal de La Laguna, 38200 Santa Cruz de Tenerife, Spain
| | | | - Araceli Delgado
- Department of Chemical Engineering and Pharmaceutical Technology, Faculty of Pharmacy, University of La Laguna, Ave. Astrofísico Fco. Sánchez s/n. San Cristóbal de La Laguna, 38200 Santa Cruz de Tenerife, Spain; (C.É.); (A.D.)
- Institute of Biomedical Technologies (ITB), Medicine Section, Faculty of Health Science, University of La Laguna, St. Santa María Soledad, s/n. San Cristóbal de La Laguna, 38200 Santa Cruz de Tenerife, Spain
| | - Teresa A. Almeida
- Department of Biochemistry, Microbiology, Cell Biology and Genetics, Biology Section, Science Faculty, University of La Laguna, Ave. Astrofísico Fco. Sánchez s/n. San Cristóbal de La Laguna, 38200 Santa Cruz de Tenerife, Spain; (A.S.); (S.B.-F.); (R.R.)
- Institute of Tropical Diseases and Healthcare of the Canary Island, Ave. Astrofísico Fco. Sánchez s/n. San Cristóbal de La Laguna, 38200 Santa Cruz de Tenerife, Spain
- Correspondence: ; Tel.: +34-922-316-502 (ext. 6117)
| |
Collapse
|
22
|
Uimari O, Subramaniam KS, Vollenhoven B, Tapmeier TT. Uterine Fibroids (Leiomyomata) and Heavy Menstrual Bleeding. FRONTIERS IN REPRODUCTIVE HEALTH 2022; 4:818243. [PMID: 36303616 PMCID: PMC9580818 DOI: 10.3389/frph.2022.818243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Uterine Fibroids, or leiomyomata, affect millions of women world-wide, with a high incidence of 75% within women of reproductive age. In ~30% of patients, uterine fibroids cause menorrhagia, or heavy menstrual bleeding, and more than half of the patients experience symptoms such as heavy menstrual bleeding, pelvic pain, or infertility. Treatment is symptomatic with limited options including hysterectomy as the most radical solution. The genetic foundations of uterine fibroid growth have been traced to somatic driver mutations (MED12, HMGA2, FH -/-, and COL4A5-A6). These also lead to downstream expression of angiogenic factors including IGF-1 and IGF-2, as opposed to the VEGF-driven mechanism found in the angiogenesis of hypoxic tumors. The resulting vasculature supplying the fibroid with nutrients and oxygen is highly irregular. Of particular interest is the formation of a pseudocapsule around intramural fibroids, a unique structure within tumor angiogenesis. These aberrations in vascular architecture and network could explain the heavy menstrual bleeding observed. However, other theories have been proposed such as venous trunks, or venous lakes caused by the blocking of normal blood flow by uterine fibroids, or the increased local action of vasoactive growth factors. Here, we review and discuss the evidence for the various hypotheses proposed.
Collapse
Affiliation(s)
- Outi Uimari
- Department of Obstetrics and Gynecology, Oulu University, Oulu, Finland
- Research Unit for Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology and Ophthalmology (PEDEGO) Research Unit and Medical Research Center Oulu, Oulu University Hospital, Oulu, Finland
| | - Kavita S. Subramaniam
- St John's Institute of Dermatology, King's College London, Guy's Hospital, London, United Kingdom
- Endometriosis CaRe Centre, Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Beverley Vollenhoven
- Women's and Newborn Program, Monash Health, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Thomas T. Tapmeier
- Endometriosis CaRe Centre, Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
23
|
Wei JJ. HMGA2: A Biomarker in Gynecologic Neoplasia. JOURNAL OF CLINICAL AND TRANSLATIONAL PATHOLOGY 2022; 2:3-7. [PMID: 35340777 PMCID: PMC8950094 DOI: 10.14218/jctp.2021.00018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
High Mobility Group A2 gene (HMGA2), an oncofetal protein, is normally expressed in fetal development and completely shuts down in almost all organs and tissue types during adulthood. It is upregulated or overexpressed again in certain mesenchymal neoplasms due to chromosomal translocations and in malignant epithelial tumors through transcription regulation. HMGA2 overexpression can either drive tumor development or promote the aggressiveness of tumor growth. Many gynecologic neoplasms, including uterine smooth muscle tumors and ovarian cancer, are associated with HMGA2 overexpression. In this article, we review recent developments in the study of HMGA2 and its expression as a potential biomarker for gynecologic neoplasms and clinic application.
Collapse
Affiliation(s)
- Jian-Jun Wei
- Correspondence to: Jian-Jun Wei, Department of Pathology, Northwestern University, School of Medicine, Feinberg 7-334, 251 East Huron Street, Chicago, IL 60611, USA. Tel: +1-312-926-1815, Fax: +1-312-926-3127,
| |
Collapse
|
24
|
Zhi S, Li J, Kong X, Xie X, Zhang Q, Fang G. Insulin-like growth factor 2 mRNA binding protein 2 regulates proliferation, migration, and angiogenesis of keratinocytes by modulating heparanase stability. Bioengineered 2021; 12:11267-11276. [PMID: 34753397 PMCID: PMC8810085 DOI: 10.1080/21655979.2021.2002495] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Wound healing is related to proliferation, migration, and angiogenesis of keratinocytes. Insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) is an important N6-methyladenosine (m6A) reader, which is involved in multiple processes, including wound healing. However, the function and mechanism of IGF2BP2 in keratinocyte processes are largely uncertain. In the present study, expression levels of IGF2BP2 and heparanase (HPSE) were detected by quantitative reverse transcription polymerase chain reaction and western blotting assays. Cell proliferation was investigated by cell counting kit-8 (CCK-8) analysis. Cell migration was determined through wound healing assay. Angiogenesis was measured by tube formation assay and vascular endothelial growth factor (VEGF) level using enzyme linked immunosorbent assay (ELISA). The interaction between IGF2BP2 and HPSE was analyzed by RNA immunoprecipitation, pull-down and luciferase reporter analyses. The results showed that IGF2BP2 expression was enhanced in wound healing. IGF2BP2 downregulation constrained HaCaT cell proliferation, migration, and angiogenesis. IGF2BP2 knockdown decreased HPSE expression. IGF2BP2 could regulate HPSE stability by binding with 3ʹ untranslated region (UTR) of HPSE. HPSE upregulation attenuated silencing IGF2BP2-mediated suppression of proliferation, migration, and angiogenesis. As a conclusion, IGF2BP2 knockdown repressed proliferation, migration, and angiogenesis of HaCaT cells by decreasing HPSE stability.
Collapse
Affiliation(s)
- Shaomin Zhi
- Department of Emergency, Xi'an No. 3 Hospital, Xi'an, Shaanxi, P.R. China
| | - Jun Li
- Department of Emergency, Xi'an No. 3 Hospital, Xi'an, Shaanxi, P.R. China
| | - Xiao Kong
- Department of Emergency, Xi'an No. 3 Hospital, Xi'an, Shaanxi, P.R. China
| | - Xuemei Xie
- Department of Emergency, Xi'an No. 3 Hospital, Xi'an, Shaanxi, P.R. China
| | - Qiangli Zhang
- Department of Emergency, Xi'an No. 3 Hospital, Xi'an, Shaanxi, P.R. China
| | - Guoxiang Fang
- Department of Emergency, Xi'an No. 3 Hospital, Xi'an, Shaanxi, P.R. China
| |
Collapse
|
25
|
Ciarmela P, Delli Carpini G, Greco S, Zannotti A, Montik N, Giannella L, Giuliani L, Grelloni C, Panfoli F, Paolucci M, Pierucci G, Ragno F, Pellegrino P, Petraglia F, Ciavattini A. Uterine fibroid vascularization: from morphological evidence to clinical implications. Reprod Biomed Online 2021; 44:281-294. [PMID: 34848152 DOI: 10.1016/j.rbmo.2021.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/24/2021] [Accepted: 09/10/2021] [Indexed: 01/04/2023]
Abstract
Uterine fibroids are the most common cause of solid pelvic tumours, occurring in 20-30% of fertile women and presenting clinical complications that seriously affect women's health. They commonly cause severe symptoms, such as heavy, prolonged menstrual bleeding and anaemia. The study of microscopic and macroscopic vascular aspects of uterine fibroids is important for understanding the clinical manifestations of uterine fibroids, for predicting the effectiveness of alternative treatments to surgery, i.e. uterine artery embolization, for improving surgery outcomes and for carrying out a differential diagnosis with other benign conditions, e.g. adenomyosis, or malignancy, e.g. leiomyosarcoma, and to develop new therapeutic approaches. In this review, current knowledge of how the vascular network and angiogenesis are implied in the formation of uterine fibroids and in the pathogenesis of related symptoms is explored, and evidence on the role of ultrasound in evaluating fibroid vascularization is summarized. This review combines anatomical, morphological and biomolecular information related to angiogenic mechanisms with diagnostic and clinical information, highlighting the various interconnections. Uterine and fibroid vascularization need further investigation to gain a deeper understanding of the pathogenetic elements that lead to the formation of uterine fibroids and their clinical manifestations.
Collapse
Affiliation(s)
- Pasquapina Ciarmela
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona 60126, Italy.
| | - Giovanni Delli Carpini
- Gynecologic Section, Department od Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Stefania Greco
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Alessandro Zannotti
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona 60126, Italy; Gynecologic Section, Department od Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Nina Montik
- Gynecologic Section, Department od Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Luca Giannella
- Gynecologic Section, Department od Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Lucia Giuliani
- Gynecologic Section, Department od Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Camilla Grelloni
- Gynecologic Section, Department od Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Francesca Panfoli
- Gynecologic Section, Department od Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Michela Paolucci
- Gynecologic Section, Department od Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Gloria Pierucci
- Gynecologic Section, Department od Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Federica Ragno
- Gynecologic Section, Department od Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Pamela Pellegrino
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Felice Petraglia
- Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and Gynecology, University of Florence, Careggi University Hospital Florence, Italy
| | - Andrea Ciavattini
- Gynecologic Section, Department od Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, Ancona 60126, Italy
| |
Collapse
|
26
|
Wu Z, Wang M, Li F, Wang F, Jia J, Feng Z, Huo X, Yang J, Jin W, Sa R, Gao W, Yu L. CDK13-Mediated Cell Cycle Disorder Promotes Tumorigenesis of High HMGA2 Expression Gastric Cancer. Front Mol Biosci 2021; 8:707295. [PMID: 34513922 PMCID: PMC8427521 DOI: 10.3389/fmolb.2021.707295] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/26/2021] [Indexed: 11/23/2022] Open
Abstract
The inhibitor of CDK4/6 has been clinically used for treating certain types of cancer which are characterized by G0/G1 acceleration induced by the CDK4/6-RB1 pathway. On the contrary, the cell cycle–related molecules are abnormal in over 50% of the patients with gastric cancer (GC), but the efficiency of inhibiting CDK4/6 does not work well as it is expected. In our study, we found HMGA2 promotes GC through accelerating the S–G2/M phase transition, instead of G0/G1. We also found CDK13 is the direct target gene of HMGA2. Importantly, we analyzed 200 pairs of GC and the adjacent tissue and proved the positive relation between HMGA2 and CDK13; moreover, high expression of both genes predicts a poorer prognosis than the expression of single gene does. We explored the effect of the novel CDK12/13 inhibiting agent, SR-4835, on high HMGA2 expression GC and found inhibition of both genes jointly could reach a satisfied result. Therefore, we suggest that inhibition of CDK13 and HMGA2 simultaneously could be an effective strategy for high HMGA2 expression GC. To detect the expression of both genes simultaneously and individually could be of benefit to predict prognosis for GC.
Collapse
Affiliation(s)
- Zhouying Wu
- Clinical Medical Research Center/Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Diseases, Inner Mongolia People's Hospital, Hohhot, China
| | - Min Wang
- Clinical Medical Research Center/Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Diseases, Inner Mongolia People's Hospital, Hohhot, China
| | - Feng Li
- Clinical Medical Research Center/Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Diseases, Inner Mongolia People's Hospital, Hohhot, China
| | - Feng Wang
- Department of Pathology, Inner Mongolia People's Hospital, Hohhot, China
| | - Jianchao Jia
- Clinical Medical Research Center/Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Diseases, Inner Mongolia People's Hospital, Hohhot, China
| | - Zongqi Feng
- Clinical Medical Research Center/Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Diseases, Inner Mongolia People's Hospital, Hohhot, China
| | - Xue Huo
- Clinical Medical Research Center/Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Diseases, Inner Mongolia People's Hospital, Hohhot, China
| | - Jie Yang
- Clinical Medical Research Center/Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Diseases, Inner Mongolia People's Hospital, Hohhot, China
| | - Wen Jin
- Clinical Medical Research Center/Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Diseases, Inner Mongolia People's Hospital, Hohhot, China
| | - Rina Sa
- Clinical Medical Research Center/Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Diseases, Inner Mongolia People's Hospital, Hohhot, China
| | - Wenming Gao
- Departments of Cardiology, Hohhot First Hospital, Hohhot, China
| | - Lan Yu
- Clinical Medical Research Center/Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Diseases, Inner Mongolia People's Hospital, Hohhot, China.,Department of Endocrine and Metabolic Diseases, Inner Mongolia People's Hospital, Hohhot, China
| |
Collapse
|
27
|
Molecular and Cellular Insights into the Development of Uterine Fibroids. Int J Mol Sci 2021; 22:ijms22168483. [PMID: 34445194 PMCID: PMC8395213 DOI: 10.3390/ijms22168483] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
Uterine leiomyomas represent the most common benign gynecologic tumor. These hormone-dependent smooth-muscle formations occur with an estimated prevalence of ~70% among women of reproductive age and cause symptoms including pain, abnormal uterine bleeding, infertility, and recurrent abortion. Despite the prevalence and public health impact of uterine leiomyomas, available treatments remain limited. Among the potential causes of leiomyomas, early hormonal exposure during periods of development may result in developmental reprogramming via epigenetic changes that persist in adulthood, leading to disease onset or progression. Recent developments in unbiased high-throughput sequencing technology enable powerful approaches to detect driver mutations, yielding new insights into the genomic instability of leiomyomas. Current data also suggest that each leiomyoma originates from the clonal expansion of a single transformed somatic stem cell of the myometrium. In this review, we propose an integrated cellular and molecular view of the origins of leiomyomas, as well as paradigm-shifting studies that will lead to better understanding and the future development of non-surgical treatments for these highly frequent tumors.
Collapse
|
28
|
Rothzerg E, Ho XD, Xu J, Wood D, Märtson A, Kõks S. Upregulation of 15 Antisense Long Non-Coding RNAs in Osteosarcoma. Genes (Basel) 2021; 12:genes12081132. [PMID: 34440306 PMCID: PMC8394133 DOI: 10.3390/genes12081132] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 12/14/2022] Open
Abstract
The human genome encodes thousands of natural antisense long noncoding RNAs (lncRNAs); they play the essential role in regulation of gene expression at multiple levels, including replication, transcription and translation. Dysregulation of antisense lncRNAs plays indispensable roles in numerous biological progress, such as tumour progression, metastasis and resistance to therapeutic agents. To date, there have been several studies analysing antisense lncRNAs expression profiles in cancer, but not enough to highlight the complexity of the disease. In this study, we investigated the expression patterns of antisense lncRNAs from osteosarcoma and healthy bone samples (24 tumour-16 bone samples) using RNA sequencing. We identified 15 antisense lncRNAs (RUSC1-AS1, TBX2-AS1, PTOV1-AS1, UBE2D3-AS1, ERCC8-AS1, ZMIZ1-AS1, RNF144A-AS1, RDH10-AS1, TRG-AS1, GSN-AS1, HMGA2-AS1, ZNF528-AS1, OTUD6B-AS1, COX10-AS1 and SLC16A1-AS1) that were upregulated in tumour samples compared to bone sample controls. Further, we performed real-time polymerase chain reaction (RT-qPCR) to validate the expressions of the antisense lncRNAs in 8 different osteosarcoma cell lines (SaOS-2, G-292, HOS, U2-OS, 143B, SJSA-1, MG-63, and MNNG/HOS) compared to hFOB (human osteoblast cell line). These differentially expressed IncRNAs can be considered biomarkers and potential therapeutic targets for osteosarcoma.
Collapse
Affiliation(s)
- Emel Rothzerg
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia; (E.R.); (J.X.); (D.W.)
- Perron Institute for Neurological and Translational Science, QEII Medical Centre, Nedlands, WA 6009, Australia
| | - Xuan Dung Ho
- Department of Oncology, College of Medicine and Pharmacy, Hue University, Hue 53000, Vietnam;
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia; (E.R.); (J.X.); (D.W.)
| | - David Wood
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia; (E.R.); (J.X.); (D.W.)
| | - Aare Märtson
- Department of Traumatology and Orthopaedics, University of Tartu, Tartu University Hospital, 50411 Tartu, Estonia;
| | - Sulev Kõks
- Perron Institute for Neurological and Translational Science, QEII Medical Centre, Nedlands, WA 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA 6150, Australia
- Correspondence: ; Tel.: +61-(0)-8-6457-0313
| |
Collapse
|
29
|
Kirschen GW, AlAshqar A, Miyashita-Ishiwata M, Reschke L, El Sabeh M, Borahay MA. Vascular biology of uterine fibroids: connecting fibroids and vascular disorders. Reproduction 2021; 162:R1-R18. [PMID: 34034234 DOI: 10.1530/rep-21-0087] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022]
Abstract
Fibroids are benign tumors caused by the proliferation of myometrial smooth muscle cells in the uterus that can lead to symptoms such as abdominal pain, constipation, urinary retention, and infertility. While traditionally thought of as a disease process intrinsic to the uterus, accumulating evidence suggests that fibroid growth may be linked with the systemic vasculature system, although cell-intrinsic factors are certainly of principal importance in their inception. Fibroids are associated with essential hypertension and preeclampsia, as well as atherosclerosis, for reasons that are becoming increasingly elucidated. Factors such as the renin-angiotensin-aldosterone system, estrogen, and endothelial dysfunction all likely play a role in fibroid pathogenesis. In this review, we lay out a framework for reconceptualizing fibroids as a systemic vascular disorder, and discuss how pharmaceutical agents and other interventions targeting the vasculature may aid in the novel treatment of fibroids.
Collapse
Affiliation(s)
- Gregory W Kirschen
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Abdelrahman AlAshqar
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Obstetrics and Gynecology, Kuwait University, Kuwait City, Kuwait
| | | | - Lauren Reschke
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Malak El Sabeh
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mostafa A Borahay
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
30
|
HMGA2 as a Critical Regulator in Cancer Development. Genes (Basel) 2021; 12:genes12020269. [PMID: 33668453 PMCID: PMC7917704 DOI: 10.3390/genes12020269] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/01/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
The high mobility group protein 2 (HMGA2) regulates gene expression by binding to AT-rich regions of DNA. Akin to other DNA architectural proteins, HMGA2 is highly expressed in embryonic stem cells during embryogenesis, while its expression is more limited at later stages of development and in adulthood. Importantly, HMGA2 is re-expressed in nearly all human malignancies, where it promotes tumorigenesis by multiple mechanisms. HMGA2 increases cancer cell proliferation by promoting cell cycle entry and inhibition of apoptosis. In addition, HMGA2 influences different DNA repair mechanisms and promotes epithelial-to-mesenchymal transition by activating signaling via the MAPK/ERK, TGFβ/Smad, PI3K/AKT/mTOR, NFkB, and STAT3 pathways. Moreover, HMGA2 supports a cancer stem cell phenotype and renders cancer cells resistant to chemotherapeutic agents. In this review, we discuss these oncogenic roles of HMGA2 in different types of cancers and propose that HMGA2 may be used for cancer diagnostic, prognostic, and therapeutic purposes.
Collapse
|
31
|
Mehine M, Khamaiseh S, Ahvenainen T, Heikkinen T, Äyräväinen A, Pakarinen P, Härkki P, Pasanen A, Bützow R, Vahteristo P. 3'RNA Sequencing Accurately Classifies Formalin-Fixed Paraffin-Embedded Uterine Leiomyomas. Cancers (Basel) 2020; 12:cancers12123839. [PMID: 33352722 PMCID: PMC7766537 DOI: 10.3390/cancers12123839] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/08/2020] [Accepted: 12/15/2020] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Uterine leiomyomas are benign smooth muscle tumors affecting millions of women globally. On a molecular level, leiomyomas can be classified into three main subtypes, each characterized by mutations affecting either MED12, HMGA2, or FH. Leiomyomas are still widely regarded as a single entity, although early observations suggest that different subtypes behave differently, in terms of both clinical outcomes and therapeutic requirements. The majority of classification studies on leiomyomas have been performed using fresh frozen tissue. Archival formalin-fixed paraffin-embedded (FFPE) tissue represents an invaluable source of biological material that can be studied retrospectively. Methods capable of generating high-quality data from FFPE material are in high demand. Here, we show that 3′RNA sequencing can accurately classify leiomyomas that have been stored as FFPE tissue in hospital archives for years. A targeted 3′RNA sequencing panel could provide researchers and clinicians with a cost-effective and scalable diagnostic tool for classifying smooth muscle tumors. Abstract Uterine leiomyomas are benign smooth muscle tumors occurring in 70% of women of reproductive age. The majority of leiomyomas harbor one of three well-established genetic changes: a hotspot mutation in MED12, overexpression of HMGA2, or biallelic loss of FH. The majority of studies have classified leiomyomas by complex and costly methods, such as whole-genome sequencing, or by combining multiple traditional methods, such as immunohistochemistry and Sanger sequencing. The type of specimens and the amount of resources available often determine the choice. A more universal, cost-effective, and scalable method for classifying leiomyomas is needed. The aim of this study was to evaluate whether RNA sequencing can accurately classify formalin-fixed paraffin-embedded (FFPE) leiomyomas. We performed 3′RNA sequencing with 44 leiomyoma and 5 myometrium FFPE samples, revealing that the samples clustered according to the mutation status of MED12, HMGA2, and FH. Furthermore, we confirmed each subtype in a publicly available fresh frozen dataset. These results indicate that a targeted 3′RNA sequencing panel could serve as a cost-effective and robust tool for stratifying both fresh frozen and FFPE leiomyomas. This study also highlights 3′RNA sequencing as a promising method for studying the abundance of unexploited tissue material that is routinely stored in hospital archives.
Collapse
Affiliation(s)
- Miika Mehine
- Applied Tumor Genomics Research Program, University of Helsinki, 00014 Helsinki, Finland; (M.M.); (S.K.); (T.A.); (T.H.); (A.Ä.); (A.P.); (R.B.)
- Department of Medical and Clinical Genetics, University of Helsinki, 00014 Helsinki, Finland
| | - Sara Khamaiseh
- Applied Tumor Genomics Research Program, University of Helsinki, 00014 Helsinki, Finland; (M.M.); (S.K.); (T.A.); (T.H.); (A.Ä.); (A.P.); (R.B.)
- Department of Medical and Clinical Genetics, University of Helsinki, 00014 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, 00014 Helsinki, Finland
| | - Terhi Ahvenainen
- Applied Tumor Genomics Research Program, University of Helsinki, 00014 Helsinki, Finland; (M.M.); (S.K.); (T.A.); (T.H.); (A.Ä.); (A.P.); (R.B.)
- Department of Medical and Clinical Genetics, University of Helsinki, 00014 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, 00014 Helsinki, Finland
| | - Tuomas Heikkinen
- Applied Tumor Genomics Research Program, University of Helsinki, 00014 Helsinki, Finland; (M.M.); (S.K.); (T.A.); (T.H.); (A.Ä.); (A.P.); (R.B.)
- Department of Medical and Clinical Genetics, University of Helsinki, 00014 Helsinki, Finland
| | - Anna Äyräväinen
- Applied Tumor Genomics Research Program, University of Helsinki, 00014 Helsinki, Finland; (M.M.); (S.K.); (T.A.); (T.H.); (A.Ä.); (A.P.); (R.B.)
- Department of Medical and Clinical Genetics, University of Helsinki, 00014 Helsinki, Finland
- Department of Obstetrics and Gynecology, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland; (P.P.); (P.H.)
| | - Päivi Pakarinen
- Department of Obstetrics and Gynecology, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland; (P.P.); (P.H.)
| | - Päivi Härkki
- Department of Obstetrics and Gynecology, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland; (P.P.); (P.H.)
| | - Annukka Pasanen
- Applied Tumor Genomics Research Program, University of Helsinki, 00014 Helsinki, Finland; (M.M.); (S.K.); (T.A.); (T.H.); (A.Ä.); (A.P.); (R.B.)
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, 00029 Helsinki, Finland
| | - Ralf Bützow
- Applied Tumor Genomics Research Program, University of Helsinki, 00014 Helsinki, Finland; (M.M.); (S.K.); (T.A.); (T.H.); (A.Ä.); (A.P.); (R.B.)
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, 00029 Helsinki, Finland
| | - Pia Vahteristo
- Applied Tumor Genomics Research Program, University of Helsinki, 00014 Helsinki, Finland; (M.M.); (S.K.); (T.A.); (T.H.); (A.Ä.); (A.P.); (R.B.)
- Department of Medical and Clinical Genetics, University of Helsinki, 00014 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, 00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-2-94125600
| |
Collapse
|
32
|
Ferrero H. HMGA2 involvement in uterine leiomyomas development through angiogenesis activation. Fertil Steril 2020; 114:974-975. [PMID: 32943224 DOI: 10.1016/j.fertnstert.2020.07.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 02/06/2023]
|