1
|
Qu L, Wang F, Wang Y, Li Z. The regulation of LRPs by miRNAs in cancer: influencing cancer characteristics and responses to treatment. Cancer Cell Int 2025; 25:182. [PMID: 40382654 PMCID: PMC12085831 DOI: 10.1186/s12935-025-03804-z] [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: 09/10/2024] [Accepted: 05/04/2025] [Indexed: 05/20/2025] Open
Abstract
The low-density lipoprotein receptor-related protein (LRP) family is a group of cell surface receptors that participate in a variety of biological processes, including lipid metabolism, Wnt signaling, and bone metabolism. miRNAs are small non-coding RNA molecules that regulate gene expression and play a role in many biological processes, including the occurrence and development of tumors. Accumulating evidence demonstrates that LRP members are modulated by miRNAs across multiple cancer types, influencing key oncogenic processes-including tumor cell proliferation, apoptosis suppression, extracellular matrix remodeling, cell adhesion, and angiogenesis. The LRPs, miRNAs, their upstream lncRNAs, and downstream signaling molecules often form complex signaling pathways to regulate the activity of tumor cells. However, the tissue-specific roles and mechanistic underpinnings of these pathways remain incompletely understood. When examining the emerging concept of the interaction between miRNAs and LRPs, we emphasize the significance of these complex regulatory layers in the initiation and progression of cancer. Collectively, these findings are critical for advancing our understanding of the role of the LRPs family in the occurrence and development of tumors, as well as for the development of new strategies for cancer treatment.
Collapse
Affiliation(s)
- Lianyue Qu
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Hospital of China Medical University, Shenyang, P. R. China
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Fan Wang
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Hospital of China Medical University, Shenyang, P. R. China
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Yuxiang Wang
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Hospital of China Medical University, Shenyang, P. R. China
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Zixuan Li
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Hospital of China Medical University, Shenyang, P. R. China.
- Department of Radiology, The First Hospital of China Medical University, Shenyang, P. R. China.
| |
Collapse
|
2
|
Wang B, Zhu Y, Liu D, Hu C, Zhu R. The intricate dance of non-coding RNAs in myasthenia gravis pathogenesis and treatment. Front Immunol 2024; 15:1342213. [PMID: 38605954 PMCID: PMC11007667 DOI: 10.3389/fimmu.2024.1342213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/11/2024] [Indexed: 04/13/2024] Open
Abstract
Myasthenia gravis (MG) stands as a perplexing autoimmune disorder affecting the neuromuscular junction, driven by a multitude of antibodies targeting postsynaptic elements. However, the mystery of MG pathogenesis has yet to be completely uncovered, and its heterogeneity also challenges diagnosis and treatment. Growing evidence shows the differential expression of non-coding RNAs (ncRNAs) in MG has played an essential role in the development of MG in recent years. Remarkably, these aberrantly expressed ncRNAs exhibit distinct profiles within diverse clinical subgroups and among patients harboring various antibody types. Furthermore, they have been implicated in orchestrating the production of inflammatory cytokines, perturbing the equilibrium of T helper 1 cells (Th1), T helper 17 cells (Th17), and regulatory T cells (Tregs), and inciting B cells to generate antibodies. Studies have elucidated that certain ncRNAs mirror the clinical severity of MG, while others may hold therapeutic significance, showcasing a propensity to return to normal levels following appropriate treatments or potentially foretelling the responsiveness to immunosuppressive therapies. Notably, the intricate interplay among these ncRNAs does not follow a linear trajectory but rather assembles into a complex network, with competing endogenous RNA (ceRNA) emerging as a prominent hub in some cases. This comprehensive review consolidates the landscape of dysregulated ncRNAs in MG, briefly delineating their pivotal role in MG pathogenesis. Furthermore, it explores their promise as prospective biomarkers, aiding in the elucidation of disease subtypes, assessment of disease severity, monitoring therapeutic responses, and as novel therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | - Ruixia Zhu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
3
|
Zhang F, Fan G, Wang X. Correlation between BTG3, CASP9 and LRP4 single-nucleotide polymorphisms and susceptibility to papillary thyroid carcinoma. Biomark Med 2022; 16:537-547. [PMID: 35362324 DOI: 10.2217/bmm-2021-0711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Objective: To study the association of BTG3, CASP9 and LRP4 single-nucleotide polymorphisms with susceptibility to papillary thyroid carcinoma (PTC). Methods: The BTG3 rs9977638, CASP9 rs884363 and LRP4 rs898604 genotypes of 175 PTC patients and 175 controls were analyzed. Results: Rs9977638 TC genotype and CC genotype, rs884363 CC genotype and rs898604 GG genotype were related to a lower PTC susceptibility risk (p < 0.01). The risk of PTC susceptibility was higher when carrying BTG3 rs9977638 CC, CASP9 rs884363 AC and LRP4 rs898604 AG at the same time (p < 0.01). Conclusion: Combined BTG3, CASP9 and LRP4 genotype analysis has a certain application value in the diagnosis of PTC.
Collapse
Affiliation(s)
- Fei Zhang
- Department of Maxillofacial & E.N.T. Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy,Tianjin Cancer Institute, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Department of Thyroid Breast Hernia Surgery, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region, 010017, China
| | - Guidong Fan
- Department of Thyroid Breast Hernia Surgery, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region, 010017, China
| | - Xudong Wang
- Department of Maxillofacial & E.N.T. Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy,Tianjin Cancer Institute, National Clinical Research Center for Cancer, Tianjin, 300060, China
| |
Collapse
|
4
|
Wang L, Zhang L. Emerging Roles of Dysregulated MicroRNAs in Myasthenia Gravis. Front Neurosci 2020; 14:507. [PMID: 32508584 PMCID: PMC7253668 DOI: 10.3389/fnins.2020.00507] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/22/2020] [Indexed: 01/03/2023] Open
Abstract
Myasthenia gravis (MG) is a rare acquired autoimmune neuromuscular disease. Autoantibodies, cellular immunity, complement, and cytokines are involved in the pathogenesis of MG. It is characterized by the dysfunction of neuromuscular junction transmission and skeletal muscle weakness. MicroRNAs (miRNAs) are non-coding small molecule ribonucleic acids that regulate various biological processes (e.g., development, differentiation, and immunity) at the transcriptional and post-transcriptional levels of gene expression. miRNAs play an important regulatory role in the pathogenesis of autoimmune diseases, including MG. In recent studies, the functional mechanisms underlying the role of miRNAs in the pathogenesis of MG have received increasing attention. miRNAs are highly stable and have high specificity in peripheral body fluids. Therefore, the miRNAs in body fluids may represent promising biomarkers for determining the prognosis of MG and the efficacy of treatment. This article reviews the role of miRNAs in the pathogenesis of MG, highlights the potential of miRNAs as new biomarkers for the diagnosis of MG, and deepens our understanding of disease processes.
Collapse
Affiliation(s)
- Lin Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lijuan Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Phylogenetic Analysis to Explore the Association Between Anti-NMDA Receptor Encephalitis and Tumors Based on microRNA Biomarkers. Biomolecules 2019; 9:biom9100572. [PMID: 31590348 PMCID: PMC6843259 DOI: 10.3390/biom9100572] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/23/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022] Open
Abstract
MicroRNA (miRNA) is a small non-coding RNA that functions in the epigenetics control of gene expression, which can be used as a useful biomarker for diseases. Anti-NMDA receptor (anti-NMDAR) encephalitis is an acute autoimmune disorder. Some patients have been found to have tumors, specifically teratomas. This disease occurs more often in females than in males. Most of them have a significant recovery after tumor resection, which shows that the tumor may induce anti-NMDAR encephalitis. In this study, I review microRNA (miRNA) biomarkers that are associated with anti-NMDAR encephalitis and related tumors, respectively. To the best of my knowledge, there has not been any research in the literature investigating the relationship between anti-NMDAR encephalitis and tumors through their miRNA biomarkers. I adopt a phylogenetic analysis to plot the phylogenetic trees of their miRNA biomarkers. From the analyzed results, it may be concluded that (i) there is a relationship between these tumors and anti-NMDAR encephalitis, and (ii) this disease occurs more often in females than in males. This sheds light on this issue through miRNA intervention.
Collapse
|
6
|
Bollard J, Patte C, Radkova K, Massoma P, Chardon L, Valantin J, Gadot N, Goddard I, Vercherat C, Hervieu V, Gouysse G, Poncet G, Scoazec JY, Walter T, Roche C. Neuropilin-2 contributes to tumor progression in preclinical models of small intestinal neuroendocrine tumors. J Pathol 2019; 249:343-355. [PMID: 31257576 DOI: 10.1002/path.5321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 05/21/2019] [Accepted: 06/21/2019] [Indexed: 12/19/2022]
Abstract
The identification of novel regulators of tumor progression is a key challenge to gain knowledge on the biology of small intestinal neuroendocrine tumors (SI-NETs). We recently identified the loss of the axon guidance protein semaphorin 3F as a protumoral event in SI-NETs. Interestingly the expression of its receptor neuropilin-2 (NRP-2) was still maintained. This study aimed at deciphering the potential role of NRP-2 as a contributor to SI-NET progression. The role of NRP-2 in SI-NET progression was addressed using an approach integrating human tissue and serum samples, cell lines and in vivo models. Data obtained from human SI-NET tissues showed that membranous NRP-2 expression is present in a majority of tumors, and is correlated with invasion, metastatic abilities, and neovascularization. In addition, NRP-2 soluble isoform was found elevated in serum samples from metastatic patients. In preclinical mouse models of NET progression, NRP-2 silencing led to a sustained antitumor effect, partly driven by the downregulation of VEGFR2. In contrast, its ectopic expression conferred a gain of aggressiveness, driven by the activation of various oncogenic signaling pathways. Lastly, NRP-2 inhibition led to a decrease of tumor cell viability, and sensitized to therapeutic agents. Overall, our results point out NRP-2 as a potential therapeutic target for SI-NETs, and will foster the development of innovative strategies targeting this receptor. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Julien Bollard
- Neuroendocrine Tumors Group, Department of Translational Research and Innovation, Centre Léon Bérard, Lyon, France.,INSERM U1052/CNRS UMR5286/University of Lyon, Cancer Research Center of Lyon, Lyon, France
| | - Céline Patte
- Neuroendocrine Tumors Group, Department of Translational Research and Innovation, Centre Léon Bérard, Lyon, France.,INSERM U1052/CNRS UMR5286/University of Lyon, Cancer Research Center of Lyon, Lyon, France
| | - Kristina Radkova
- Neuroendocrine Tumors Group, Department of Translational Research and Innovation, Centre Léon Bérard, Lyon, France.,INSERM U1052/CNRS UMR5286/University of Lyon, Cancer Research Center of Lyon, Lyon, France
| | - Patrick Massoma
- INSERM U1052/CNRS UMR5286/University of Lyon, Cancer Research Center of Lyon, Lyon, France
| | - Laurence Chardon
- Department of Biology and Hormonology, Lyon-Est Hospital, Bron, France
| | - Julie Valantin
- Pathology-Research Platform, Department of Translational Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Nicolas Gadot
- Pathology-Research Platform, Department of Translational Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Isabelle Goddard
- Laboratoire des Modèles Tumoraux, Lyon Synergie Cancer, Lyon, France
| | - Cécile Vercherat
- Neuroendocrine Tumors Group, Department of Translational Research and Innovation, Centre Léon Bérard, Lyon, France.,INSERM U1052/CNRS UMR5286/University of Lyon, Cancer Research Center of Lyon, Lyon, France
| | - Valérie Hervieu
- Neuroendocrine Tumors Group, Department of Translational Research and Innovation, Centre Léon Bérard, Lyon, France.,INSERM U1052/CNRS UMR5286/University of Lyon, Cancer Research Center of Lyon, Lyon, France.,Department of Pathology, Lyon-Est Hospital, Bron, France
| | | | - Gilles Poncet
- Neuroendocrine Tumors Group, Department of Translational Research and Innovation, Centre Léon Bérard, Lyon, France.,INSERM U1052/CNRS UMR5286/University of Lyon, Cancer Research Center of Lyon, Lyon, France.,Department of Medical Oncology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Jean-Yves Scoazec
- Department of Pathology, Gustave-Roussy Cancer Campus, Villejuif, France
| | - Thomas Walter
- Neuroendocrine Tumors Group, Department of Translational Research and Innovation, Centre Léon Bérard, Lyon, France.,INSERM U1052/CNRS UMR5286/University of Lyon, Cancer Research Center of Lyon, Lyon, France.,Department of Medical Oncology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Colette Roche
- Neuroendocrine Tumors Group, Department of Translational Research and Innovation, Centre Léon Bérard, Lyon, France.,INSERM U1052/CNRS UMR5286/University of Lyon, Cancer Research Center of Lyon, Lyon, France
| |
Collapse
|
7
|
Scarpa A. The landscape of molecular alterations in pancreatic and small intestinal neuroendocrine tumours. ANNALES D'ENDOCRINOLOGIE 2019; 80:153-158. [PMID: 31072588 DOI: 10.1016/j.ando.2019.04.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gastroenteropancreatic Neuroendocrine Neoplasms (GEP-NENs) arise throughout the gut and feature varying biological behaviour and malignant potential. GEP-NENs include two genetically different entities, well-differentiated neuroendocrine tumours (NETs) and poorly differentiated neuroendocrine carcinomas (NEC). NECs are characterized by a dismal prognosis and by distinctive TP53 and RB1 inactivation which sets them apart from NETs. The latter, conversely, have a wide spectrum of aggressiveness and molecular alterations. Knowledge on their biology has recently expanded thanks to high-throughput studies focused on two important groups of well-differentiated neuroendocrine neoplasms: pancreatic (PanNETs) and small intestinal (SiNETs) tumours. PanNETs have been among the most studied also due to genetic syndromes featuring their onset. Research stemming from this observation has uncovered the inactivation of MEN1, VHL, TSC1/2, and the hyperactivation of the PI3K/mTOR pathway as distinctive biological features of these neoplasms. Next-Generation Sequencing added information on the role of telomere lengthening via ATRX/DAXX inactivation in a fraction of PanNETs, while other display shortened telomeres and recurrent chromosomal alterations. The data so far disclosed a heterogeneous combination of driver events, yet converging into four pathways including DNA damage repair, cell cycle regulation, PI3K/mTOR signalling and telomere maintenance. SiNETs showed a lesser relationship with mutational driver events, even in the case of familial cases. High throughput studies identified putative driver mutations in CDKN1 and APC which, however, were reported in a minor fraction (∼10%) of cases. Tumorigenesis of SiNETs seems to depend more on chromosomal alterations (loss of chromosome 8, gains at 4, 5 and 20) and epigenetic events, which converge to hyperactivate the PI3K/mTOR, MAPK and Wnt pathways. While calling for further integrative studies, these data lay previous and recent findings in a more defined frame and provide clinical research with several candidate markers for patient stratification and companion diagnostics.
Collapse
Affiliation(s)
- Aldo Scarpa
- RC-Net Centre for applied research on cancer, University and Hospital Trust of Verona, 37134 Verona, Italy; Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy.
| |
Collapse
|
8
|
Mafficini A, Scarpa A. Genetics and Epigenetics of Gastroenteropancreatic Neuroendocrine Neoplasms. Endocr Rev 2019; 40:506-536. [PMID: 30657883 PMCID: PMC6534496 DOI: 10.1210/er.2018-00160] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 12/27/2018] [Indexed: 12/11/2022]
Abstract
Gastroenteropancreatic (GEP) neuroendocrine neoplasms (NENs) are heterogeneous regarding site of origin, biological behavior, and malignant potential. There has been a rapid increase in data publication during the last 10 years, mainly driven by high-throughput studies on pancreatic and small intestinal neuroendocrine tumors (NETs). This review summarizes the present knowledge on genetic and epigenetic alterations. We integrated the available information from each compartment to give a pathway-based overview. This provided a summary of the critical alterations sustaining neoplastic cells. It also highlighted similarities and differences across anatomical locations and points that need further investigation. GEP-NENs include well-differentiated NETs and poorly differentiated neuroendocrine carcinomas (NECs). NENs are graded as G1, G2, or G3 based on mitotic count and/or Ki-67 labeling index, NECs are G3 by definition. The distinction between NETs and NECs is also linked to their genetic background, as TP53 and RB1 inactivation in NECs set them apart from NETs. A large number of genetic and epigenetic alterations have been reported. Recurrent changes have been traced back to a reduced number of core pathways, including DNA damage repair, cell cycle regulation, and phosphatidylinositol 3-kinase/mammalian target of rapamycin signaling. In pancreatic tumors, chromatin remodeling/histone methylation and telomere alteration are also affected. However, also owing to the paucity of disease models, further research is necessary to fully integrate and functionalize data on deregulated pathways to recapitulate the large heterogeneity of behaviors displayed by these tumors. This is expected to impact diagnostics, prognostic stratification, and planning of personalized therapy.
Collapse
Affiliation(s)
- Andrea Mafficini
- ARC-Net Center for Applied Research on Cancer, University and Hospital Trust of Verona, Verona, Italy.,Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Aldo Scarpa
- ARC-Net Center for Applied Research on Cancer, University and Hospital Trust of Verona, Verona, Italy.,Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| |
Collapse
|
9
|
Zhou X, Xia E, Bhandari A, Zheng C, Xiang J, Guan Y, Zhang X. LRP4 promotes proliferation, migration, and invasion in papillary thyroid cancer. Biochem Biophys Res Commun 2018; 503:257-263. [PMID: 29885843 DOI: 10.1016/j.bbrc.2018.06.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 06/06/2018] [Indexed: 01/10/2023]
Abstract
Dysregulation of cell proliferation and death is considered the foundation of the malignant biological characteristics of cancer. In this study, we conducted a comprehensive analysis of a massively parallel whole transcriptome resequencing of paired papillary thyroid cancer and normal thyroid tissues from 19 patients. In addition, we found that LRP4, a member of the low-density lipoprotein receptor-related protein family, is significantly overexpressed in thyroid carcinoma. We demonstrated through quantitative real-time polymerase chain reaction (qRT-PCR) that LRP4 is upregulated in papillary thyroid cancer (PTC) tissues. This observation was also consistent with data analyzed from The Cancer Genome Atlas (TCGA) cohort. Thus, the biological role of LRP4 in the thyroid cancer in the present study was investigated using the PTC cell lines TPC1, BCPAP and KTC-1. In these cell lines, the mRNA level of LRP4 was higher than normal thyroid cancer cell named HTORI3. In vitro experiments demonstrated that LRP4 downregulation significantly inhibited the colony formation, proliferation, migration, and invasion of the three PTC cell lines. Knockdown of LRP4 by small interfering RNA (siRNA) in those cell lines decreased the protein expression of N-cadherin, Enhancer of zeste homolog 2 (EZH2), and Zinc finger E-box-binding home-box 1 (ZEB1). Furthermore, LRP4 knockdown significantly reduced the levels of phosphorylated PI3K in the PTC cell lines. In conclusion, the present study indicated that LRP4 is a gene associated with PTC and might become a potential therapeutic target.
Collapse
Affiliation(s)
- Xiaofen Zhou
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Erjie Xia
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Adheesh Bhandari
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Chen Zheng
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Jingjing Xiang
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Yaoyao Guan
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Xiaohua Zhang
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China.
| |
Collapse
|
10
|
Monazzam A, Lau J, Velikyan I, Li SC, Razmara M, Rosenström U, Eriksson O, Skogseid B. Increased Expression of GLP-1R in Proliferating Islets of Men1 Mice is Detectable by [ 68Ga]Ga-DO3A-VS-Cys 40-Exendin-4 /PET. Sci Rep 2018; 8:748. [PMID: 29335487 PMCID: PMC5768696 DOI: 10.1038/s41598-017-18855-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 12/13/2017] [Indexed: 12/22/2022] Open
Abstract
Multiple endocrine neoplasia type 1 (MEN1) is an endocrine tumor syndrome caused by heterozygous mutations in the MEN1 tumor suppressor gene. The MEN1 pancreas of the adolescent gene carrier frequently contain diffusely spread pre-neoplasias and microadenomas, progressing to macroscopic and potentially malignant pancreatic neuroendocrine tumors (P-NET), which represents the major death cause in MEN1. The unveiling of the molecular mechanism of P-NET which is not currently understood fully to allow the optimization of diagnostics and treatment. Glucagon-like peptide 1 (GLP-1) pathway is essential in islet regeneration, i.e. inhibition of β-cell apoptosis and enhancement of β-cell proliferation, yet involvement of GLP-1 in MEN1 related P-NET has not yet been demonstrated. The objective of this work was to investigate if normal sized islets of Men1 heterozygous mice have increased Glucagon-like peptide-1 receptor (GLP-1R) expression compared to wild type islets, and if this increase is detectable in vivo with positron emission tomography (PET) using [68Ga]Ga-DO3A-VS-Cys40-Exendin-4 (68Ga-Exendin-4). 68Ga-Exendin-4 showed potential for early lesion detection in MEN1 pancreas due to increased GLP1R expression.
Collapse
Affiliation(s)
- Azita Monazzam
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Joey Lau
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Irina Velikyan
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Su-Chen Li
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Masoud Razmara
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Ulrika Rosenström
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Olof Eriksson
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Britt Skogseid
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
11
|
Malczewska A, Kidd M, Matar S, Kos-Kudla B, Modlin IM. A Comprehensive Assessment of the Role of miRNAs as Biomarkers in Gastroenteropancreatic Neuroendocrine Tumors. Neuroendocrinology 2018; 107:73-90. [PMID: 29566385 DOI: 10.1159/000487326] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/25/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND/AIMS A key issue in neuroendocrine neoplasia management is the identification of blood signatures that specifically define the activity of a cancer or local tumor microenvironment. MicroRNAs (miRNAs) may represent such a candidate. To evaluate their clinical utility as biomarkers in gastroenteropancreatic neuroendocrine tumors (GEP-NETs), we assessed their expression in tissue and blood. METHODS A systematic review of PubMed was undertaken to identify studies investigating miRNAs in GEP-NETs and their utility as blood or tissue biomarkers. RESULTS Twenty-two studies using a range of methodologies with different normalization protocols were identified: tumor - gastric NET type 1 (n = 1 study: MiR-222, regulates p27KIP1), pancreatic (n = 6: MiR-21 [inflammatory marker, oncogene] and MiR-144 [PI3K/AKT signaling], both up- and downregulated depending on the method), small intestinal (n = 7: no consistent signature), and colorectal (n = 3: no consistent signature); blood - gastric NET type 1 (n = 1: MiR-222), pancreatic (n = 3: MiR-21), and small intestinal (n = 3: no consistent signature). The studies all included heterogeneous cohorts, were insufficiently powered, and utilized different methodologies, and age- and gender-matched controls were not used. Different miRNA isolation methods and detection protocols resulted in inconsistent expression comparing tumor and blood. A scientific discrepancy was the downregulated expression of some circulating candidates compared to tissue levels, suggesting methodological issues or physiological responses to the tumor. Both are of concern in defining the biometrics of a marker. CONCLUSIONS A potential biomarker for GEP-NETs included MiR-21 (small bowel and pancreas), but this epithelial tumor marker requires prospective validation. Overall, significant scientific investigation remains to identify and demonstrate neuroendocrine specificity and to validate candidate miRNA biomarkers.
Collapse
Affiliation(s)
- Anna Malczewska
- Department of Endocrinology and Neuroendocrine Tumors, Medical University of Silesia, Katowice, Poland
| | - Mark Kidd
- Wren Laboratories, Branford, Connecticut, USA
| | - Somer Matar
- Wren Laboratories, Branford, Connecticut, USA
| | - Beata Kos-Kudla
- Department of Endocrinology and Neuroendocrine Tumors, Medical University of Silesia, Katowice, Poland
| | - Irvin M Modlin
- Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
12
|
Finnerty BM, Gray KD, Moore MD, Zarnegar R, Fahey III TJ. Epigenetics of gastroenteropancreatic neuroendocrine tumors: A clinicopathologic perspective. World J Gastrointest Oncol 2017; 9:341-353. [PMID: 28979716 PMCID: PMC5605334 DOI: 10.4251/wjgo.v9.i9.341] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 06/27/2017] [Accepted: 08/04/2017] [Indexed: 02/05/2023] Open
Abstract
Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are a heterogeneous group of rare tumors whose site-specific tumor incidence and clinical behavior vary widely. Genetic alterations associated with familial inherited syndromes have been well defined; however, the genetic profile of sporadic tumors is less clear as their tumorigenesis does not appear to be controlled by classic oncogenes such as P53, RB, or KRAS. Even within GEP-NETs, there are no common oncogenic drivers; for example, DAXX/ATRX mutations are strongly implicated in the tumorigenesis of pancreatic but not small bowel NETs. Accordingly, the dysregulation of epigenetic mechanisms has been hypothesized as a potential regulator of GEP-NET tumorigenesis and has become a major focus of recent studies. Despite the heterogeneity of tumor cohorts evaluated in these studies, it is obvious that there are methylation patterns, chromatin remodeling alterations, and microRNA and long non-coding RNA (lncRNA) differential expression profiles that are distinctive of GEP-NETs, some of which are correlated with significant differences in clinical outcomes. Several translational studies have provided convincing data identifying potential prognostic biomarkers, and some of these have demonstrated preliminary success as serum biomarkers that can be used clinically. Nevertheless, there are many opportunities to further define the mechanisms by which these epigenetic modifications influence tumorigenesis, and this will provide better insight into their prognostic and therapeutic utility. Furthermore, these findings form the foundation for future studies evaluating the clinical efficacy of epigenetic modifications as prognostic biomarkers, as well as potential therapeutic targets.
Collapse
Affiliation(s)
- Brendan M Finnerty
- Department of Surgery, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, United States
| | - Katherine D Gray
- Department of Surgery, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, United States
| | - Maureen D Moore
- Department of Surgery, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, United States
| | - Rasa Zarnegar
- Department of Surgery, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, United States
| | - Thomas J Fahey III
- Department of Surgery, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, United States
| |
Collapse
|
13
|
Xu H, Li G, Yue Z, Li C. HCV core protein-induced upregulation of microRNA-196a promotes aberrant proliferation in hepatocellular carcinoma by targeting FOXO1. Mol Med Rep 2016; 13:5223-9. [PMID: 27108614 DOI: 10.3892/mmr.2016.5159] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 03/11/2016] [Indexed: 11/06/2022] Open
Abstract
The hepatitis C virus (HCV) core protein is critical in the development of hepatocellular carcinoma (HCC). Investigations on HCC have previously focused on microRNAs, a class of small non‑coding RNAs, which are crucial in cancer development and progression. The present study aimed to investigate whether microRNA (miR)‑196a is aberrantly regulated by the HCV core protein, and whether miR‑196a is involved in the regulation of the aberrant proliferation of HCV‑HCC cells. In the study, miRNA expression was detected by quantitative polymerase chain reaction analysis. An Ad‑HCV core adenovirus was constructed and cell proliferation was measured using a Cell Counting Kit-8 assay and a cell cycle assay following infection. The results of the present study demonstrated that the HCV core protein increased the expression of miR‑196a, and that overexpression of miR‑196a in the HepG2 and Huh‑7 HCC cell lines promoted cell proliferation by inducing the G1‑S transition. Furthermore, the present study demonstrated that forkhead box O1 (FOXO1) was directly regulated by miR‑196a, and was essential in mediating the biological effects of miR‑196a in HCC. The overexpression of FOXO1 markedly reversed the effect of miR‑196a in HCC cell proliferation. Taken together, the data obtained in the present study provided compelling evidence that elevated expression levels of miR‑196a by the HCV core protein can function as an onco‑microRNA during HCV‑induced cell proliferation by downregulating the expression of FOXO1, indicating a potential novel therapeutic target for HCV-related HCC.
Collapse
Affiliation(s)
- Hao Xu
- Department of Infectious Diseases, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Guangming Li
- Department of Hepatology, The 6th People's Hospital of Zhengzhou, Zhengzhou, Henan 450000, P.R. China
| | - Zhanyi Yue
- Department of Laboratory Diagnosis, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Chengzhong Li
- Department of Infectious Diseases, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| |
Collapse
|