1
|
Wang J, Ke N, Wu X, Zhen H, Hu J, Liu X, Li S, Zhao F, Li M, Shi B, Zhao Z, Ren C, Hao Z. MicroRNA-148a Targets DNMT1 and PPARGC1A to Regulate the Viability, Proliferation, and Milk Fat Synthesis of Ovine Mammary Epithelial Cells. Int J Mol Sci 2024; 25:8558. [PMID: 39201245 PMCID: PMC11354201 DOI: 10.3390/ijms25168558] [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: 06/06/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 09/02/2024] Open
Abstract
In this study, the expression profiles of miR-148a were constructed in eight different ovine tissues, including mammary gland tissue, during six different developmental periods. The effect of miR-148a on the viability, proliferation, and milk fat synthesis of ovine mammary epithelial cells (OMECs) was investigated, and the target relationship of miR-148a with two predicted target genes was verified. The expression of miR-148a exhibited obvious tissue-specific and temporal-specific patterns. miR-148a was expressed in all eight ovine tissues investigated, with the highest expression level in mammary gland tissue (p < 0.05). Additionally, miR-148a was expressed in ovine mammary gland tissue during each of the six developmental periods studied, with its highest level at peak lactation (p < 0.05). The overexpression of miR-148a increased the viability of OMECs, the number and percentage of Edu-labeled positive OMECs, and the expression levels of two cell-proliferation marker genes. miR-148a also increased the percentage of OMECs in the S phase. In contrast, transfection with an miR-148a inhibitor produced the opposite effect compared to the miR-148a mimic. These results indicate that miR-148a promotes the viability and proliferation of OMECs in Small-tailed Han sheep. The miR-148a mimic increased the triglyceride content by 37.78% (p < 0.01) and the expression levels of three milk fat synthesis marker genes in OMECs. However, the miR-148a inhibitor reduced the triglyceride level by 87.11% (p < 0.01). These results suggest that miR-148a promotes milk fat synthesis in OMECs. The dual-luciferase reporter assay showed that miR-148a reduced the luciferase activities of DNA methyltransferase 1 (DNMT1) and peroxisome proliferator-activated receptor gamma coactivator 1-A (PPARGC1A) in wild-type vectors, suggesting that they are target genes of miR-148a. The expression of miR-148a was highly negatively correlated with PPARGC1A (r = -0.789, p < 0.001) in ovine mammary gland tissue, while it had a moderate negative correlation with DNMT1 (r = -0.515, p = 0.029). This is the first study to reveal the molecular mechanisms of miR-148a underlying the viability, proliferation, and milk fat synthesis of OMECs in sheep.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Zhiyun Hao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (J.W.); (N.K.); (X.W.); (H.Z.); (J.H.); (X.L.); (S.L.); (F.Z.); (M.L.); (B.S.); (Z.Z.); (C.R.)
| |
Collapse
|
2
|
Schirripa A, Sexl V, Kollmann K. Cyclin-dependent kinase inhibitors in malignant hematopoiesis. Front Oncol 2022; 12:916682. [PMID: 36033505 PMCID: PMC9403899 DOI: 10.3389/fonc.2022.916682] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
The cell-cycle is a tightly orchestrated process where sequential steps guarantee cellular growth linked to a correct DNA replication. The entire cell division is controlled by cyclin-dependent kinases (CDKs). CDK activation is balanced by the activating cyclins and CDK inhibitors whose correct expression, accumulation and degradation schedule the time-flow through the cell cycle phases. Dysregulation of the cell cycle regulatory proteins causes the loss of a controlled cell division and is inevitably linked to neoplastic transformation. Due to their function as cell-cycle brakes, CDK inhibitors are considered as tumor suppressors. The CDK inhibitors p16INK4a and p15INK4b are among the most frequently altered genes in cancer, including hematopoietic malignancies. Aberrant cell cycle regulation in hematopoietic stem cells (HSCs) bears severe consequences on hematopoiesis and provokes hematological disorders with a broad array of symptoms. In this review, we focus on the importance and prevalence of deregulated CDK inhibitors in hematological malignancies.
Collapse
|
3
|
Shi L, Feng L, Tong Y, Jia J, Li T, Wang J, Jiang Z, Yu M, Xia H, Jin Q, Jiang X, Cheng Y, Ju L, Liu J, Zhang Q, Lou J. Genome wide profiling of miRNAs relevant to the DNA damage response induced by hexavalent chromium exposure (DDR-related miRNAs in response to Cr (VI) exposure). ENVIRONMENT INTERNATIONAL 2021; 157:106782. [PMID: 34329887 DOI: 10.1016/j.envint.2021.106782] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 06/13/2023]
Abstract
AIM We aimed to explore the expression of miRNAs and their potential roles in the DNA damage response (DDR) induced by Cr (VI) exposure in human B lymphoblast cells (HMy2.CIR cells) and in a population of Cr (VI)-exposed humans. METHODS Differentially expressed miRNAs were found by a combination of miRNA sequencing and RT-qPCR validation in HMy2.CIR cells treated with K2Cr2O7. Differentially expressed miRNAs related to DDR were selected for functional study. The expression levels of differential miRNAs were also investigated in chromate workers. RESULTS A total of 214 differentially expressed miRNAs were identified by sequencing, and the expression of 5 miRNAs among 25 associated with DDR was found to be consistent between sequencing and validation studies.Functional studies showed that miR-148a-3p, miR-21-5p, and miR-424-3p might be related to Cr (VI)-induced cell apoptosis, and miR-221-3p might participate in Cr (VI)-induced DDR. We also found that the expression of miR-21-5p and miR-424-3p was upregulated in chromate workers. CONCLUSIONS Cr (VI) exposure could significantly impact miRNAs expression in vitro and in chromate workers. Functional studies showed that miR-148a-3p, miR-21-5p and miR-221-3p might take a crucial role in the cellular DDR induced by Cr (VI) exposure.
Collapse
Affiliation(s)
- Li Shi
- School of Public Health (Institute of Occupational Diseases), Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, China
| | - Lingfang Feng
- School of Public Health (Institute of Occupational Diseases), Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, China
| | - Yan Tong
- Affiliated Hangzhou First People's Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China
| | - Junlin Jia
- Center for Biostatistics, Bioinformatics and Big Data, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tao Li
- School of Public Health (Institute of Occupational Diseases), Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, China
| | - Jing Wang
- School of Public Health (Institute of Occupational Diseases), Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, China
| | - Zhaoqiang Jiang
- School of Public Health (Institute of Occupational Diseases), Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, China
| | - Min Yu
- School of Public Health (Institute of Occupational Diseases), Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, China
| | - Hailing Xia
- School of Public Health (Institute of Occupational Diseases), Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, China
| | - Qi Jin
- School of Public Health (Institute of Occupational Diseases), Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, China
| | - Xiyi Jiang
- School of Public Health (Institute of Occupational Diseases), Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, China
| | - Yongran Cheng
- School of Public Health (Institute of Occupational Diseases), Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, China
| | - Li Ju
- School of Public Health (Institute of Occupational Diseases), Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, China
| | - Jiaqi Liu
- School of Public Health (Institute of Occupational Diseases), Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, China
| | - Quan Zhang
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Jianlin Lou
- School of Public Health (Institute of Occupational Diseases), Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Xiang P, Yeung YT, Wang J, Wu Q, Du R, Huang C, Jia X, Gao Y, Zhi Y, Guo F, Wei H, Zhang D, Liu Y, Liu L, Liang L, Wang J, Song Y, Liu K, Fang B. miR-17-3p promotes the proliferation of multiple myeloma cells by downregulating P21 expression through LMLN inhibition. Int J Cancer 2021; 148:3071-3085. [PMID: 33609405 PMCID: PMC8248421 DOI: 10.1002/ijc.33528] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/31/2021] [Accepted: 02/11/2021] [Indexed: 01/01/2023]
Abstract
Multiple myeloma (MM), a hematological malignancy, has a poor prognosis and requires an invasive procedure. Reports have implicated miRNAs in the diagnosis, treatment and prognosis of hematological malignancies. In our study, we evaluated the expression profiles of miR-17-3p in plasma and bone marrow mononuclear cells of monoclonal gammopathy of undetermined significance (MGUS) and MM patients and healthy subjects. The results showed that the plasma and mononuclear cell expression levels of miR-17-3p in MM patients were higher than those in MGUS patients and normal controls. In addition, the expression of miR-17-3p was positively correlated with diagnostic indexes, such as marrow plasma cell abundance and serum M protein level, and positively correlated with the International Staging System stage of the disease. Receiver operating characteristic curve analysis suggested that miR-17-3p might be a diagnostic index of MM. Moreover, miR-17-3p regulated cell proliferation, apoptosis and the cell cycle through P21 in MM cell lines and promoted MM tumor growth in vivo. Furthermore, we predicted and verified LMLN as a functional downstream target gene of miR-17-3p. Negatively regulated by miR-17-3p, LMLN inhibits MM cell growth, exerting a tumor suppressive function through P21. Taken together, our data identify miR-17-3p as a promising diagnostic biomarker for MM in the clinic and unveil a new miR-17-3p-LMLN-P21 axis in MM progression.
Collapse
Affiliation(s)
- Pu Xiang
- Department of HematologyAffiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Henan Hematology InstituteZhengzhouHenanChina
| | - Yiu To Yeung
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
| | - Jiheng Wang
- Department of Head and Neck ThyroidAffiliated Cancer Hospital of Zhengzhou University and Henan Cancer HospitalZhengzhouHenanChina
| | - Qiong Wu
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Ruijuan Du
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Chuntian Huang
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Xuechao Jia
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Yunfeng Gao
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
| | - Yafei Zhi
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Fangqin Guo
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Huifang Wei
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Dandan Zhang
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
| | - Yuzhang Liu
- Department of HematologyAffiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Henan Hematology InstituteZhengzhouHenanChina
| | - Lina Liu
- Department of HematologyAffiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Henan Hematology InstituteZhengzhouHenanChina
| | - Lijie Liang
- Department of HematologyAffiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Henan Hematology InstituteZhengzhouHenanChina
| | - Juan Wang
- Department of HematologyAffiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Henan Hematology InstituteZhengzhouHenanChina
| | - Yongping Song
- Department of HematologyAffiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Henan Hematology InstituteZhengzhouHenanChina
| | - Kangdong Liu
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Cancer Chemoprevention International Collaboration LaboratoryZhengzhouHenanChina
| | - Baijun Fang
- Department of HematologyAffiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Henan Hematology InstituteZhengzhouHenanChina
| |
Collapse
|
5
|
Tellier J. miR-148a weaves its thread into the plasma cell fate. Eur J Immunol 2021; 51:1076-1079. [PMID: 33792033 DOI: 10.1002/eji.202149240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 03/16/2021] [Accepted: 03/24/2021] [Indexed: 12/22/2022]
Abstract
The plasma cells (PC) are characterized by their rarity, their formidable capacity to continuously secrete massive amounts of antibodies and the potential to live through the whole life span of the organism that houses them. Because of the potency of their effector function, their differentiation and survival are tightly regulated. The PC identity is implemented and maintained by a transcriptional program that allow them to face the challenges entailed by their longevity and high metabolic activity. The main transcription factors overseeing this transcriptional network have been identified (BLIMP1, IRF4, XBP1), but new players, like miRNA, continue to emerge and bring new layers of complexity to the regulatory loops. In the current issue of the European Journal of Immunology [Eur. J. Immunol. 2021. 51: 1089-1109], Pracht et al. identify miR-148a as a significant actor of the PC program that favors the differentiation through the inhibition of competitor fates, and supports the survival and fitness of the long-lived PC. In this commentary, we will discuss the place of miR-148a in the PC transcriptional network and its potential as a therapeutic target in PC-driven diseases.
Collapse
Affiliation(s)
- Julie Tellier
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
6
|
Pracht K, Meinzinger J, Schulz SR, Daum P, Côrte-Real J, Hauke M, Roth E, Kindermann D, Mielenz D, Schuh W, Wittmann J, Jäck HM. miR-148a controls metabolic programming and survival of mature CD19-negative plasma cells in mice. Eur J Immunol 2021; 51:1089-1109. [PMID: 33336366 DOI: 10.1002/eji.202048993] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/08/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022]
Abstract
Long-lived antibody-secreting plasma cells are essential to establish humoral memory against pathogens. While a regulatory transcription factor network has been established in plasma cell differentiation, the regulatory role of miRNAs remains enigmatic. We have recently identified miR-148a as the most abundant miRNA in primary mouse and human plasma cells. To determine whether this plasma cell signature miRNA controls the in vivo development of B cells into long-lived plasma cells, we established mice with genomic, conditional, and inducible deletions of miR-148a. The analysis of miR-148a-deficient mice revealed reduced serum Ig, decreased numbers of newly formed plasmablasts and reduced CD19-negative, CD93-positive long-lived plasma cells. Transcriptome and metabolic analysis revealed an impaired glucose uptake, a reduced oxidative phosphorylation-based energy metabolism, and an altered abundance of homing receptors CXCR3 (increase) and CXCR4 (reduction) in miR-148a-deficient plasma cells. These findings support the role of miR-148a as a positive regulator of the maintenance of long-lived plasma cells.
Collapse
Affiliation(s)
- Katharina Pracht
- Division of Molecular Immunology, Internal Medicine III, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Meinzinger
- Division of Molecular Immunology, Internal Medicine III, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian R Schulz
- Division of Molecular Immunology, Internal Medicine III, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Patrick Daum
- Division of Molecular Immunology, Internal Medicine III, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Joana Côrte-Real
- Division of Molecular Immunology, Internal Medicine III, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Manuela Hauke
- Division of Molecular Immunology, Internal Medicine III, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Edith Roth
- Division of Molecular Immunology, Internal Medicine III, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Dorothea Kindermann
- Division of Molecular Immunology, Internal Medicine III, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Dirk Mielenz
- Division of Molecular Immunology, Internal Medicine III, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Wolfgang Schuh
- Division of Molecular Immunology, Internal Medicine III, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Jürgen Wittmann
- Division of Molecular Immunology, Internal Medicine III, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Internal Medicine III, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
7
|
Razavipour SF, Harikumar KB, Slingerland JM. p27 as a Transcriptional Regulator: New Roles in Development and Cancer. Cancer Res 2020; 80:3451-3458. [PMID: 32341036 DOI: 10.1158/0008-5472.can-19-3663] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/25/2020] [Accepted: 04/21/2020] [Indexed: 11/16/2022]
Abstract
p27 binds and inhibits cyclin-CDK to arrest the cell cycle. p27 also regulates other processes including cell migration and development independent of its cyclin-dependent kinase (CDK) inhibitory action. p27 is an atypical tumor suppressor-deletion or mutational inactivation of the gene encoding p27, CDKN1B, is rare in human cancers. p27 is rarely fully lost in cancers because it can play both tumor suppressive and oncogenic roles. Until recently, the paradigm was that oncogenic deregulation results from either loss of growth restraint due to excess p27 proteolysis or from an oncogenic gain of function through PI3K-mediated C-terminal p27 phosphorylation, which disrupts the cytoskeleton to increase cell motility and metastasis. In cancers, C-terminal phosphorylation alters p27 protein-protein interactions and shifts p27 from CDK inhibitor to oncogene. Recent data indicate p27 regulates transcription and acts as a transcriptional coregulator of cJun. C-terminal p27 phosphorylation increases p27-cJun recruitment to and action on target genes to drive oncogenic pathways and repress differentiation programs. This review focuses on noncanonical, CDK-independent functions of p27 in migration, invasion, development, and gene expression, with emphasis on how transcriptional regulation by p27 illuminates its actions in cancer. A better understanding of how p27-associated transcriptional complexes are regulated might identify new therapeutic targets at the interface between differentiation and growth control.
Collapse
Affiliation(s)
- Seyedeh Fatemeh Razavipour
- Breast Cancer Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC
| | - Kuzhuvelil B Harikumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| | - Joyce M Slingerland
- Breast Cancer Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC.
| |
Collapse
|
8
|
Abstract
Antibody-secreting plasma cells are the central pillars of humoral immunity. They are generated in a fundamental cellular restructuring process from naive B cells upon contact with antigen. This outstanding process is guided and controlled by a complex transcriptional network accompanied by a fascinating morphological metamorphosis, governed by the combined action of Blimp-1, Xbp-1 and IRF-4. The survival of plasma cells requires the intimate interaction with a specific microenvironment, consisting of stromal cells and cells of hematopoietic origin. Cell-cell contacts, cytokines and availability of metabolites such as glucose and amino acids modulate the survival abilities of plasma cells in their niches. Moreover, plasma cells have been shown to regulate immune responses by releasing cytokines. Furthermore, plasma cells are central players in autoimmune diseases and malignant transformation of plasma cells can result in the generation of multiple myeloma. Hence, the development of sophisticated strategies to deplete autoreactive plasma cells and myeloma cells represents a challenge for current and future research.
Collapse
Affiliation(s)
- Wolfgang Schuh
- Division of Molecular Immunology, Department of Internal Medicine III, Nikolaus-Fiebiger Center, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany.
| | - Dirk Mielenz
- Division of Molecular Immunology, Department of Internal Medicine III, Nikolaus-Fiebiger Center, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine III, Nikolaus-Fiebiger Center, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
9
|
Ferroportin downregulation promotes cell proliferation by modulating the Nrf2-miR-17-5p axis in multiple myeloma. Cell Death Dis 2019; 10:624. [PMID: 31423010 PMCID: PMC6698482 DOI: 10.1038/s41419-019-1854-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 07/19/2019] [Accepted: 07/26/2019] [Indexed: 12/20/2022]
Abstract
Recent findings demonstrate that aberrant downregulation of the iron-exporter protein, ferroportin (FPN1), is associated with poor prognosis and osteoclast differentiation in multiple myeloma (MM). Here, we show that FPN1 was downregulated in MM and that clustered regularly interspaced short palindromic repeat (CRISPR)-mediated FPN1 knockout promoted MM cell growth and survival. Using a microRNA target-scan algorithm, we identified miR-17-5p as an FPN1 regulator that promoted cell proliferation and cell cycle progression, and inhibited apoptosis—both in vitro and in vivo. miR-17-5p inhibited retarded tumor growth in a MM xenograft model. Moreover, restoring FPN1 expression at least partially abrogated the biological effects of miR-17-5p in MM cells. The cellular iron concentration regulated the expression of the iron-regulatory protein (IRP) via the 5′-untranslated region of IRP messenger RNA and modulated the post-transcriptional stability of FPN1. Bioinformatics analysis with subsequent chromatin immunoprecipitation-polymerase chain reaction and luciferase activity experiments revealed that the transcription factor Nrf2 drove FPN1 transcription through promoter binding and suppressed miR-17-5p (which also increased FPN1 expression). Nrf2-mediated FPN1 downregulation promoted intracellular iron accumulation and reactive oxygen species. Our study links FPN1 transcriptional and post-transcriptional regulation with MM cell growth and survival, and validates the prognostic value of FPN1 and its utility as a novel therapeutic target in MM.
Collapse
|
10
|
Friedrich M, Pracht K, Mashreghi MF, Jäck HM, Radbruch A, Seliger B. The role of the miR-148/-152 family in physiology and disease. Eur J Immunol 2017; 47:2026-2038. [DOI: 10.1002/eji.201747132] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/30/2017] [Accepted: 09/01/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Michael Friedrich
- Institute of Medical Immunology; Martin-Luther-University Halle-Wittenberg; Halle/Saale Germany
| | - Katharina Pracht
- Division of Molecular Immunology; Nikolaus-Fiebiger Center; Department of Internal Medicine III; University of Erlangen-Nürnberg; Erlangen Germany
| | | | - Hans-Martin Jäck
- Division of Molecular Immunology; Nikolaus-Fiebiger Center; Department of Internal Medicine III; University of Erlangen-Nürnberg; Erlangen Germany
| | | | - Barbara Seliger
- Institute of Medical Immunology; Martin-Luther-University Halle-Wittenberg; Halle/Saale Germany
| |
Collapse
|
11
|
Yang N, Chen J, Zhang H, Wang X, Yao H, Peng Y, Zhang W. LncRNA OIP5-AS1 loss-induced microRNA-410 accumulation regulates cell proliferation and apoptosis by targeting KLF10 via activating PTEN/PI3K/AKT pathway in multiple myeloma. Cell Death Dis 2017; 8:e2975. [PMID: 28796257 PMCID: PMC5596549 DOI: 10.1038/cddis.2017.358] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/15/2017] [Accepted: 06/23/2017] [Indexed: 12/16/2022]
Abstract
Numerous studies confirmed that aberrant miRNAs expression contributes to multiple myeloma (MM) development and progression. However, the roles of specific miRNAs in MM remain to be investigated. In present study, we demonstrated that miR-410 expression was increased in MM newly diagnosed and relapsed tissues and cell lines. Clinical analysis revealed that miR-410 was positively correlated with advanced ISS stage. Moreover, high miR-410 expression in MM patients showed an obvious shorter overall survival and progression-free survival. Gain- and loss-of function experiments indicated that miR-410 promoted cell proliferation, cell cycle progression and apoptosis inhibition both in vitro and in vivo. Moreover, KLF10 was identified as a direct downstream target of miR-410 in MM cells, and mediated the functional influence of miR-410 in MM, resulting in PTEN/AKT activation. In clinical samples of MM, miR-410 inversely correlated with KLF10. Alteration of KLF10 expression or AKT inhibitor at least partially abolished the biological effects of miR-410 on MM cells. Furthermore, downregulated expression of lncRNA OIP5-AS1 was inversely correlated with miR-410 expression in MM tissues. LncRNA OIP5-AS1 could modulate the miR-410 expression and regulate its target KLF10/PTEN/AKT-mediated cellular behaviors. Taken together, this research supports the first evidence that lncRNA OIP5-AS1 loss-induced miR-410 accumulation facilitates cell proliferation, cycle progression and apoptosis inhibition by targeting KLF10 via activating PTEN/PI3K/AKT pathway in MM.
Collapse
Affiliation(s)
- Nan Yang
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, West Five Road, NO.157, Xi’an, China
| | - Jinqiu Chen
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, West Five Road, NO.157, Xi’an, China
| | - Hui Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, West Five Road, NO.157, Xi’an, China
| | - Xiaman Wang
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, West Five Road, NO.157, Xi’an, China
| | - Huan Yao
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, West Five Road, NO.157, Xi’an, China
| | - Yue Peng
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, West Five Road, NO.157, Xi’an, China
| | - Wanggang Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, West Five Road, NO.157, Xi’an, China
| |
Collapse
|
12
|
Alles J, Ludwig N, Rheinheimer S, Leidinger P, Grässer FA, Keller A, Meese E. MiR-148a impairs Ras/ERK signaling in B lymphocytes by targeting SOS proteins. Oncotarget 2017; 8:56417-56427. [PMID: 28915601 PMCID: PMC5593572 DOI: 10.18632/oncotarget.17662] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 04/24/2017] [Indexed: 12/12/2022] Open
Abstract
Although microRNAs have been recognized as central cellular regulators, there is an evident lack of knowledge about their targets. Here, we analyzed potential target genes for miR-148a functioning in Ras signaling in B cells, including SOS1 and SOS2. A dual-luciferase reporter assay showed significantly decreased luciferase activity upon ectopic overexpression of miR-148a in HEK-293T cells that were co-transfected with the 3′UTR of either SOS1 or SOS2. Each of the 3′UTRs of SOS1 and SOS2 contained two binding sites for miR-148a both of which were necessary for the decreased luciferase activity. MiR-148a overexpression in HEK-293T lead to significantly reduced levels of both endogenous SOS1 and SOS2 proteins. Likewise, reduced levels of SOS proteins were found in two B cell lines that were transfected with miR-148a. The level of ERK1/2 phosphorylation as one of the most relevant downstream members of the Ras/ERK signaling pathway was also reduced in cells with miR-148a overexpression. The data show that miR-148a impairs the Ras/ERK signaling pathway via SOS1 and SOS2 proteins in B cells.
Collapse
Affiliation(s)
- Julia Alles
- Institute of Human Genetics, Saarland University, 66421 Homburg, Germany
| | - Nicole Ludwig
- Institute of Human Genetics, Saarland University, 66421 Homburg, Germany
| | | | - Petra Leidinger
- Institute of Human Genetics, Saarland University, 66421 Homburg, Germany
| | | | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, 66123 Saarbrücken, Germany
| | - Eckart Meese
- Institute of Human Genetics, Saarland University, 66421 Homburg, Germany
| |
Collapse
|
13
|
Chen L, Yang J, Xing Z, Yuan F, Shu Y, Zhang Y, Kong X, Huang T, Li H, Cai YD. An integrated method for the identification of novel genes related to oral cancer. PLoS One 2017; 12:e0175185. [PMID: 28384236 PMCID: PMC5383255 DOI: 10.1371/journal.pone.0175185] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/21/2017] [Indexed: 12/18/2022] Open
Abstract
Cancer is a significant public health problem worldwide. Complete identification of genes related to one type of cancer facilitates earlier diagnosis and effective treatments. In this study, two widely used algorithms, the random walk with restart algorithm and the shortest path algorithm, were adopted to construct two parameterized computational methods, namely, an RWR-based method and an SP-based method; based on these methods, an integrated method was constructed for identifying novel disease genes. To validate the utility of the integrated method, data for oral cancer were used, on which the RWR-based and SP-based methods were trained, thereby building two optimal methods. The integrated method combining these optimal methods was further adopted to identify the novel genes of oral cancer. As a result, 85 novel genes were inferred, among which eleven genes (e.g., MYD88, FGFR2, NF-κBIA) were identified by both the RWR-based and SP-based methods, 70 genes (e.g., BMP4, IFNG, KITLG) were discovered only by the RWR-based method and four genes (L1R1, MCM6, NOG and CXCR3) were predicted only by the SP-based method. Extensive analyses indicate that several novel genes have strong associations with cancers, indicating the effectiveness of the integrated method for identifying disease genes.
Collapse
Affiliation(s)
- Lei Chen
- School of Life Sciences, Shanghai University, Shanghai, People’s Republic of China
- College of Information Engineering, Shanghai Maritime University, Shanghai, People’s Republic of China
| | - Jing Yang
- School of Life Sciences, Shanghai University, Shanghai, People’s Republic of China
| | - Zhihao Xing
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Fei Yuan
- Department of Science & Technology, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Yang Shu
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - YunHua Zhang
- School of Resources and Environment, Anhui Agricultural University, Hefei, Anhui, People’s Republic of China
| | - XiangYin Kong
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People’s Republic of China
- * E-mail: (TH); (HPL); (YDC)
| | - HaiPeng Li
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People’s Republic of China
- * E-mail: (TH); (HPL); (YDC)
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, People’s Republic of China
- * E-mail: (TH); (HPL); (YDC)
| |
Collapse
|