1
|
Bahram Sangani N, Koetsier J, Gomes AR, Diogo MM, Fernandes TG, Bouwman FG, Mariman ECM, Ghazvini M, Gribnau J, Curfs LMG, Reutelingsperger CP, Eijssen LMT. Involvement of extracellular vesicle microRNA clusters in developing healthy and Rett syndrome brain organoids. Cell Mol Life Sci 2024; 81:410. [PMID: 39305343 PMCID: PMC11416455 DOI: 10.1007/s00018-024-05409-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/19/2024] [Accepted: 08/10/2024] [Indexed: 09/25/2024]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by de novo mutations in the MECP2 gene. Although miRNAs in extracellular vesicles (EVs) have been suggested to play an essential role in several neurological conditions, no prior study has utilized brain organoids to profile EV-derived miRNAs during normal and RTT-affected neuronal development. Here we report the spatiotemporal expression pattern of EV-derived miRNAs in region-specific forebrain organoids generated from female hiPSCs with a MeCP2:R255X mutation and the corresponding isogenic control. EV miRNA and protein expression profiles were characterized at day 0, day 13, day 40, and day 75. Several members of the hsa-miR-302/367 cluster were identified as having a time-dependent expression profile with RTT-specific alterations at the latest developmental stage. Moreover, the miRNA species of the chromosome 14 miRNA cluster (C14MC) exhibited strong upregulation in RTT forebrain organoids irrespective of their spatiotemporal location. Together, our results suggest essential roles of the C14MC and hsa-miR-302/367 clusters in EVs during normal and RTT-associated neurodevelopment, displaying promising prospects as biomarkers for monitoring RTT progression.
Collapse
Affiliation(s)
- Nasim Bahram Sangani
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands
| | - Jarno Koetsier
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands
| | - Ana Rita Gomes
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Margarida Diogo
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Tiago G Fernandes
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Freek G Bouwman
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Edwin C M Mariman
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Mehrnaz Ghazvini
- Erasmus MC iPS Facility, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Joost Gribnau
- Erasmus MC iPS Facility, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
- Department of Developmental Biology, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Leopold M G Curfs
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands
| | - Chris P Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands.
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands.
| | - Lars M T Eijssen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
- Department of Bioinformatics-BiGCaT, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
| |
Collapse
|
2
|
Chu X, Tian W, Ning J, Xiao G, Zhou Y, Wang Z, Zhai Z, Tanzhu G, Yang J, Zhou R. Cancer stem cells: advances in knowledge and implications for cancer therapy. Signal Transduct Target Ther 2024; 9:170. [PMID: 38965243 PMCID: PMC11224386 DOI: 10.1038/s41392-024-01851-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/27/2024] [Accepted: 04/28/2024] [Indexed: 07/06/2024] Open
Abstract
Cancer stem cells (CSCs), a small subset of cells in tumors that are characterized by self-renewal and continuous proliferation, lead to tumorigenesis, metastasis, and maintain tumor heterogeneity. Cancer continues to be a significant global disease burden. In the past, surgery, radiotherapy, and chemotherapy were the main cancer treatments. The technology of cancer treatments continues to develop and advance, and the emergence of targeted therapy, and immunotherapy provides more options for patients to a certain extent. However, the limitations of efficacy and treatment resistance are still inevitable. Our review begins with a brief introduction of the historical discoveries, original hypotheses, and pathways that regulate CSCs, such as WNT/β-Catenin, hedgehog, Notch, NF-κB, JAK/STAT, TGF-β, PI3K/AKT, PPAR pathway, and their crosstalk. We focus on the role of CSCs in various therapeutic outcomes and resistance, including how the treatments affect the content of CSCs and the alteration of related molecules, CSCs-mediated therapeutic resistance, and the clinical value of targeting CSCs in patients with refractory, progressed or advanced tumors. In summary, CSCs affect therapeutic efficacy, and the treatment method of targeting CSCs is still difficult to determine. Clarifying regulatory mechanisms and targeting biomarkers of CSCs is currently the mainstream idea.
Collapse
Affiliation(s)
- Xianjing Chu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wentao Tian
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jiaoyang Ning
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Gang Xiao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yunqi Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ziqi Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhuofan Zhai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Guilong Tanzhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Jie Yang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China.
| |
Collapse
|
3
|
Xu M, Xu L. Up-Regulation of miR-26a-5p Promoted Cell Growth and Tumor Metastasis of Intracranial Malignancy Through Phosphatase and Tensin Homolog Deleted on Chromosome Ten/Phosphatidylinositol3-Kinase/Protein Kinase B Signaling Pathway. J BIOMATER TISS ENG 2023. [DOI: 10.1166/jbt.2023.3228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Objective: Intracranial malignancy has ranked the 6th and 3rd in terms of global tumor morbidity and mortality, respectively. MicroRNA (miRNA) can regulate the cell physiological process. Methods: In previous study, we explored the anti-cancer effects and mechanism of
miR-26a-5p in human glioma. MiR-26a-5p expression was increased in patient with glioma. Up-regulation of miR-26a-5p promoted cell growth and tumor metastasis of human glioma through inactivation of PTEN/PI3K/Akt. Results: Down-regulation of miR-26a-5p reduced cell growth and tumor metastasis
of human glioma. Downregulation of miR-26a-5p induced PTEN protein expression, and reduced PI3K and p-Akt protein expression in human glioma. PTEN or PI3K inhibitor reduced the effects of miR-26a-5p on cell growth and tumor metastasis of human glioma. Conclusion: Our findings proved
that the cancer effect of MiR-26a-5p regulates PTEN expression and promoted cell growth of human glioma through PI3K/Akt signalling pathway.
Collapse
Affiliation(s)
- Mingtao Xu
- Department of Neurosurgery, Huhhot First Hospital, Hohhot, 010030, China
| | - Li Xu
- Department of Cardiovascular Medicine, The Affiliated People’s Hospital of Inner Mongolia Medical University, Hohhot, 010010, China
| |
Collapse
|
4
|
Rajabi A, Kayedi M, Rahimi S, Dashti F, Mirazimi SMA, Homayoonfal M, Mahdian SMA, Hamblin MR, Tamtaji OR, Afrasiabi A, Jafari A, Mirzaei H. Non-coding RNAs and glioma: Focus on cancer stem cells. Mol Ther Oncolytics 2022; 27:100-123. [PMID: 36321132 PMCID: PMC9593299 DOI: 10.1016/j.omto.2022.09.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Glioblastoma and gliomas can have a wide range of histopathologic subtypes. These heterogeneous histologic phenotypes originate from tumor cells with the distinct functions of tumorigenesis and self-renewal, called glioma stem cells (GSCs). GSCs are characterized based on multi-layered epigenetic mechanisms, which control the expression of many genes. This epigenetic regulatory mechanism is often based on functional non-coding RNAs (ncRNAs). ncRNAs have become increasingly important in the pathogenesis of human cancer and work as oncogenes or tumor suppressors to regulate carcinogenesis and progression. These RNAs by being involved in chromatin remodeling and modification, transcriptional regulation, and alternative splicing of pre-mRNA, as well as mRNA stability and protein translation, play a key role in tumor development and progression. Numerous studies have been performed to try to understand the dysregulation pattern of these ncRNAs in tumors and cancer stem cells (CSCs), which show robust differentiation and self-regeneration capacity. This review provides recent findings on the role of ncRNAs in glioma development and progression, particularly their effects on CSCs, thus accelerating the clinical implementation of ncRNAs as promising tumor biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Ali Rajabi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mehrdad Kayedi
- Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shiva Rahimi
- School of Medicine,Fasa University of Medical Sciences, Fasa, Iran
| | - Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mina Homayoonfal
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mohammad Amin Mahdian
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Omid Reza Tamtaji
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Afrasiabi
- Department of Internal Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ameneh Jafari
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
5
|
Li J, Gu J, Wang J, You A, Zhang Y, Rao G, Li S, Ge X, Zhang K, Wang D. MicroRNA-433-3p enhances chemosensitivity of glioma to cisplatin by downregulating NR5A2. Brain Behav 2022; 12:e2632. [PMID: 36303447 PMCID: PMC9759127 DOI: 10.1002/brb3.2632] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/21/2022] [Accepted: 04/24/2022] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE We attempted to investigate influence of microRNA-433-3p on malignant progression of glioma and identify its molecular mechanism, thus laying groundwork for glioma management. METHODS Expression data along with clinical data of glioma were accessed from the TCGA database for differential and survival analyses to look for the target differentially expressed genes. Quantitative reverse transcriptase PCR (qRT-PCR) and western blot were utilized to assess NR5A2 mRNA and protein expression in different glioma cell lines, respectively. MTT, Transwell assay, and flow cytometry were carried out to assay the impact of NR5A2 on behaviors of glioma cells in vitro. Bioinformatics analysis was used to identify the upstream microRNA of NR5A2 in glioma, while dual-luciferase and western blot assays were used to detect binding of microRNA and NR5A2. Chemosensitivity of glioma cells was evaluated by cisplatin cytotoxicity test. RESULTS NR5A2 was upregulated in both glioma tissues and cell lines. Dual-luciferase assay result showed binding site of microRNA-433-3p on NR5A2 mRNA 3'UTR, and microRNA-433-3p reduced NR5A2 expression. Cell assays revealed that silencing NR5A2 could hamper proliferation, invasion, and migration and enhance chemosensitivity to cisplatin while promoting glioma cell apoptosis and blocking glioma cells in G0/G1 phase. Rescue experiments also indicated that microRNA-433-3p suppressed glioma malignant progression via inhibiting NR5A2. CONCLUSION MicroRNA-433-3p which is significantly poorly expressed in glioma targets NR5A2 to suppress glioma malignant progression and enhance chemosensitivity to cisplatin.
Collapse
Affiliation(s)
- Jun Li
- The Fourth Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, China
| | - Jingshun Gu
- The Fourth Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, China
| | - Juntong Wang
- The Fourth Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, China
| | - Aiwu You
- The Fourth Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, China
| | - Yuyan Zhang
- The Fourth Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, China
| | - Guomin Rao
- The Fourth Department of Neurology, Tangshan Gongren Hospital, Tangshan, China
| | - Shuang Li
- Department of Traditional Chinese Medicine, Tangshan Gongren Hospital, Tangshan, China
| | - Xuehua Ge
- The Fourth Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, China
| | - Kun Zhang
- The Fourth Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, China
| | - Dongchun Wang
- The Fourth Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, China
| |
Collapse
|
6
|
Mafi A, Rahmati A, Babaei Aghdam Z, Salami R, Salami M, Vakili O, Aghadavod E. Recent insights into the microRNA-dependent modulation of gliomas from pathogenesis to diagnosis and treatment. Cell Mol Biol Lett 2022; 27:65. [PMID: 35922753 PMCID: PMC9347108 DOI: 10.1186/s11658-022-00354-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/22/2022] [Indexed: 11/11/2022] Open
Abstract
Gliomas are the most lethal primary brain tumors in adults. These highly invasive tumors have poor 5-year survival for patients. Gliomas are principally characterized by rapid diffusion as well as high levels of cellular heterogeneity. However, to date, the exact pathogenic mechanisms, contributing to gliomas remain ambiguous. MicroRNAs (miRNAs), as small noncoding RNAs of about 20 nucleotides in length, are known as chief modulators of different biological processes at both transcriptional and posttranscriptional levels. More recently, it has been revealed that these noncoding RNA molecules have essential roles in tumorigenesis and progression of multiple cancers, including gliomas. Interestingly, miRNAs are able to modulate diverse cancer-related processes such as cell proliferation and apoptosis, invasion and migration, differentiation and stemness, angiogenesis, and drug resistance; thus, impaired miRNAs may result in deterioration of gliomas. Additionally, miRNAs can be secreted into cerebrospinal fluid (CSF), as well as the bloodstream, and transported between normal and tumor cells freely or by exosomes, converting them into potential diagnostic and/or prognostic biomarkers for gliomas. They would also be great therapeutic agents, especially if they could cross the blood–brain barrier (BBB). Accordingly, in the current review, the contribution of miRNAs to glioma pathogenesis is first discussed, then their glioma-related diagnostic/prognostic and therapeutic potential is highlighted briefly.
Collapse
Affiliation(s)
- Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Atefe Rahmati
- Department of Hematology and Blood Banking, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Basic Science, Neyshabur University of Medical Science, Neyshabur, Iran
| | - Zahra Babaei Aghdam
- Imaging Sciences Research Group, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raziyeh Salami
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Marziyeh Salami
- Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Esmat Aghadavod
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran. .,Department of Clinical Biochemistry, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
7
|
Cai Y, An B, Yao D, Zhou H, Zhu J. MicroRNA miR-30a inhibits cisplatin resistance in ovarian cancer cells through autophagy. Bioengineered 2021; 12:10713-10722. [PMID: 34747309 PMCID: PMC8810079 DOI: 10.1080/21655979.2021.2001989] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
We study whether microRNA miR-30a inhibits the autophagy through transforming growth factor (TGF)-β/Smad4 to generate cisplatin (DDP) resistance in ovarian cancer cells. The expression of miR-30a, Smad4, and TGF-β was detected in the serum of ovarian cancer patients and DDP-resistant cell lines (A2780) by quantitative real-time polymerase chain reaction (qRT-PCR). Computational search and western blotting were used to demonstrate the downstream target of miR-30a in ovarian cancer cells. Cell viability was measured with CCK8 assay. Apoptosis and autophagy of ovarian cancer cells were analyzed by flow cytometry and transmission electron microscopy, and the expressions of Beclin1 and LC3II protein were detected by western blotting. Expression of miR-30a was significantly decreased, while expressions of TGF-β and Smad4 mRNA were increased in serum of ovarian cancer patients after DDP chemotherapy as well as in DDP-resistant cells. Activation of autophagy contributed to DDP-resistance cells. Moreover, Bioinformatics software predicted Smad4 to be a target of miR-30a. Overexpression of miR-30a decreased the expression of Smad4 and TGF-β. Additionally, miR-30a-overexpressing inhibited DDP-induce autophagy and promoted DDP-resistant cell apoptosis. In conclusion, miR-30a mediates DDP resistance in ovarian cancer by inhibiting autophagy via the TGF-β/Smad4 pathway.
Collapse
Affiliation(s)
- Yi Cai
- Department of Oncology, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Baiping An
- Department of Oncology, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dejiao Yao
- Department of Oncology, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Zhou
- Department of Oncology, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Zhu
- Department of Oncology, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
8
|
Alcedo KP, Bowser JL, Snider NT. The elegant complexity of mammalian ecto-5'-nucleotidase (CD73). Trends Cell Biol 2021; 31:829-842. [PMID: 34116887 PMCID: PMC8448938 DOI: 10.1016/j.tcb.2021.05.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 12/14/2022]
Abstract
Purinergic signaling is a fundamental mechanism used by all cells to control their internal activities and interact with the environment. A key component of the purinergic system, the enzyme ecto-5'-nucleotidase (CD73) catalyzes the last step in the extracellular metabolism of ATP to form adenosine. Efforts to harness the therapeutic potential of endogenous adenosine in cancer have culminated in the ongoing clinical development of multiple CD73-targeting antibodies and small-molecule inhibitors. However, recent studies are painting an increasingly complex picture of CD73 mRNA and protein regulation and function in cellular homeostasis, physiological adaptation, and disease development. This review discusses the latest conceptual and methodological advances that are helping to unravel the complexity of this important enzyme that was identified nearly 90 years ago.
Collapse
Affiliation(s)
- Karel P Alcedo
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jessica L Bowser
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Natasha T Snider
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
9
|
Jia J, Ouyang Z, Wang M, Ma W, Liu M, Zhang M, Yu M. MicroRNA-361-5p slows down gliomas development through regulating UBR5 to elevate ATMIN protein expression. Cell Death Dis 2021; 12:746. [PMID: 34321465 PMCID: PMC8319180 DOI: 10.1038/s41419-021-04010-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 04/21/2021] [Accepted: 05/13/2021] [Indexed: 12/13/2022]
Abstract
MicroRNA (miR)-361-5p has been studied to suppress gliomas development. Based on that, an insight into the regulatory mechanism of miR-361-5p in gliomas was supplemented from ubiquitin protein ligase E3 component N-recognin 5 (UBR5)-mediated ubiquitination of ataxia-telangiectasia mutated interactor (ATMIN). miR-361-5p, ATMIN, and UBR5 levels were clinically analyzed in gliomas tissues, which were further validated in gliomas cell lines. Loss/gain-of-function method was applied to determine the roles of miR-361-5p and UBR5 in gliomas, as to cell viability, migration, invasion, colony formation ability, and apoptosis in vitro and tumorigenesis in vivo. The relationship between miR-361-5p and UBR5 was verified and the interaction between UBR5 and ATMIN was explored. It was detected that reduced miR-361-5p and ATMIN and enhanced UBR5 levels showed in gliomas. Elevating miR-361-5p was repressive in gliomas progression. UBR5 was directly targeted by miR-361-5p. UBR5 can ubiquitinate ATMIN. miR-361-5p suppressed gliomas by regulating UBR5-mediated ubiquitination of ATMIN. Downregulating UBR5 impeded gliomas tumor growth in vivo. Upregulating miR-361-5p targets UBR5 to promote ATMIN protein expression, thus to recline the malignant phenotype of gliomas cells.
Collapse
Affiliation(s)
- Jiaoying Jia
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Zhu Ouyang
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Ming Wang
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Wenjia Ma
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Min Liu
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Mingming Zhang
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
| | - Mengqiang Yu
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
10
|
Comertpay B, Gulfidan G, Arga KY, Gov E. Cancer Stem Cell Transcriptome Profiling Reveals Seed Genes of Tumorigenesis: New Avenues for Cancer Precision Medicine. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:372-388. [PMID: 34037481 DOI: 10.1089/omi.2021.0021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer stem-like cells (CSCs) possess the ability to self-renew and differentiate, and they are among the major factors driving tumorigenesis, metastasis, and resistance to chemotherapy. Therefore, it is critical to understand the molecular substrates of CSC biology so as to discover novel molecular biosignatures that distinguish CSCs and tumor cells. Here, we report new findings and insights by employing four transcriptome datasets associated with CSCs, with CSC and tumor samples from breast, lung, oral, and ovarian tissues. The CSC samples were analyzed to identify differentially expressed genes between CSC and tumor phenotypes. Through comparative profiling of expression levels in different cancer types, we identified 17 "seed genes" that showed a mutual differential expression pattern. We showed that these seed genes were strongly associated with cancer-associated signaling pathways and biological processes, the immune system, and the key cancer hallmarks. Further, the seed genes presented significant changes in their expression profiles in different cancer types and diverse mutation rates, and they also demonstrated high potential as diagnostic and prognostic biomarkers in various cancers. We report a number of seed genes that represent significant potential as "systems biomarkers" for understanding the pathobiology of tumorigenesis. Seed genes offer a new innovation avenue for potential applications toward cancer precision medicine in a broad range of cancers in oncology in the future.
Collapse
Affiliation(s)
- Betul Comertpay
- Department of Bioengineering, Faculty of Engineering, Adana Alparslan Türkeş Science and Technology University, Adana, Turkey
| | - Gizem Gulfidan
- Department of Bioengineering, Marmara University, Istanbul, Turkey
| | | | - Esra Gov
- Department of Bioengineering, Faculty of Engineering, Adana Alparslan Türkeş Science and Technology University, Adana, Turkey
| |
Collapse
|
11
|
Chen G, Chen Z, Zhao H. MicroRNA-155-3p promotes glioma progression and temozolomide resistance by targeting Six1. J Cell Mol Med 2020; 24:5363-5374. [PMID: 32220051 PMCID: PMC7205810 DOI: 10.1111/jcmm.15192] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/29/2020] [Accepted: 03/09/2020] [Indexed: 12/13/2022] Open
Abstract
The prognosis of glioma is generally poor and is the cause of primary malignancy in the brain. The role of microRNAs has been implicated in tumour inhibition or activation. In several cancers, the Six1 signalling pathway has been found to be aberrant and also relates to the formation of tumours. We analysed the database for expression profiles and clinical specimens of various grades of glioma to assess microRNA-155-3p (miR-155-3p) expression. The role of miR-155-3p in glioblastoma, cell cycle, proliferation, apoptosis and resistance to temozolomide was assessed in vitro through flow cytometry and cell proliferation assays. Bioinformatics analyses, and assays using luciferase reporter, and immunoblotting revealed that miR-155-3p targets Six1 and that the relationship between glioma and healthy brain tissues was significantly inverse. In rescue experiments, overexpressed Six1 revoked the changes in cell cycle distribution, proliferation and resistance to temozolomide estimated by apoptosis induced by overexpressed miR-155-3p. MiR-155-3p inhibition reduced glioma cell growth and proliferation in the brain of a mouse model and increased the survival of mice with gliomas. Thus, miR-155-3p modulates Six1 expression and facilitates the progression of glioblastoma and resistance to temozolomide and may act as a novel diagnostic biomarker and a target for glioma treatment.
Collapse
Affiliation(s)
- Guangyong Chen
- Neurosurgery DepartmentChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Zhuo Chen
- Neurosurgery DepartmentChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Hang Zhao
- Neurosurgery DepartmentChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| |
Collapse
|