1
|
Stanisławowski M. Effect of adipose tissue on the development of multiple myeloma. Mol Biol Rep 2024; 52:74. [PMID: 39708277 DOI: 10.1007/s11033-024-10174-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
Multiple myeloma (MM), also referred to as Kahler's disease, is a cancer characterized by the uncontrolled growth of abnormal plasma cells and is associated with alterations in the bone tissue microenvironment. Bone marrow adipose tissue (BMAT), which comprises approximately ten percent of total body fat, can influence the progression, survival, and drug resistance of MM cells through paracrine, hormonal, and metabolic pathways. Obesity can lead to an increase in BMAT mass, which not only disrupts bone metabolism but also reduces bone density, potentially progressing from monoclonal gammopathy of undetermined significance, a benign condition, to MM. A range of factors, including impaired fatty acid metabolism, increased production of adipokines that support myeloma, and heightened expression of oncogenic microRNAs in multiple myeloma, contribute to the progression of this incurable blood cancer. To better understand the relationship between excess adipose tissue accumulation and the risk of developing multiple myeloma, a comprehensive review of published data was conducted.
Collapse
Affiliation(s)
- Marcin Stanisławowski
- Department of Histology, Medical University of Gdańsk, Dębinki 1, 80-211, Gdańsk, Poland.
| |
Collapse
|
2
|
Sogbein O, Paul P, Umar M, Chaari A, Batuman V, Upadhyay R. Bortezomib in cancer therapy: Mechanisms, side effects, and future proteasome inhibitors. Life Sci 2024; 358:123125. [PMID: 39413903 DOI: 10.1016/j.lfs.2024.123125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/07/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
The ubiquitin-proteasome pathway (UPP) regulates protein stability and normal cellular functions with the help of autocatalytic proteasome complex. Studies have linked aberrant proteasome activity to malignant cells and found that proteasome inhibitors play a significant role as therapeutic drugs for various types of cancer, specifically multiple myeloma and mantle cell lymphoma. Bortezomib, the first FDA-approved proteasome inhibitor for treating different stages of multiple myeloma, acts on cancer cells by inhibiting the 26S proteasome, modulating NF-κB, phosphorylating Bcl-2, upregulating of NOXA, blocking p53 degradation, activating caspase, generating reactive oxygen species (ROS), and inhibiting angiogenesis. However, its efficacy is limited due to side effects such as peripheral neuropathy (PN), thrombotic microangiopathy (TMA), and acute interstitial nephritis (AIN). Therefore, a better understanding of its precise mechanism of action may help mitigate these side effects. In this review, we have discussed the proposed mechanisms of action and off target effects of Bortezomib, along with the prospects of next generation potential proteasome inhibitor drugs in the treatment of cancer.
Collapse
Affiliation(s)
- Olusola Sogbein
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Pradipta Paul
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Qatar
| | - Meenakshi Umar
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ali Chaari
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Qatar
| | - Vecihi Batuman
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Rohit Upadhyay
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
3
|
Wu Z, Xia M, Wang J, Aguilar MM, Buist-Homan M, Moshage H. Extracellular vesicles originating from steatotic hepatocytes promote hepatic stellate cell senescence via AKT/mTOR signaling. Cell Biochem Funct 2024; 42:e4077. [PMID: 38881228 DOI: 10.1002/cbf.4077] [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: 02/16/2024] [Revised: 05/24/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024]
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is increasing rapidly due to the obesity epidemic. In the inflammatory stages of MASLD (MASH), activation of hepatic stellate cells (HSCs) leads to initiation and progression of liver fibrosis. Extracellular vesicles (EVs) are released from all cell types and play an important role in intercellular communication. However, the role of EVs released from hepatocytes in the context of MASLD is largely unknown. Therefore, the present study aimed to investigate the role of EVs derived from both normal and steatotic (free fatty acid-treated) hepatocytes on the phenotype of HSCs via the senescence pathway. Primary rat hepatocytes were treated with free fatty acids (FFAs: oleic acid and palmitic acid). EVs were collected by ultracentrifugation. EVs markers and HSCs activation and senescence markers were assessed by Western blot analysis, qPCR and cytochemistry. Reactive oxygen species (ROS) production was assessed by fluorescence assay. RNA profiles of EVs were evaluated by sequencing. We found that EVs from hepatocytes treated with FFAs (FFA-EVs) inhibit collagen type 1 and α-smooth muscle actin expression, increase the production of ROS and the expression of senescence markers (IL-6, IL-1β, p21 and senescence-associated β-galactosidase activity) in early activating HSCs via the AKT-mTOR pathway. Sequencing showed differentially enriched RNA species between the EVs groups. In conclusion, EVs from FFA-treated hepatocytes inhibit HSC activation by inducing senescence via the AKT-mTOR signaling pathway. Determining the components in EVs from steatotic hepatocytes that induce HSC senescence may lead to the identification of novel targets for intervention in the treatment of MASLD in the future.
Collapse
Affiliation(s)
- Zongmei Wu
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Mengmeng Xia
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Junyu Wang
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Magnolia Martinez Aguilar
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Manon Buist-Homan
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
4
|
The Multiple Myeloma Landscape: Epigenetics and Non-Coding RNAs. Cancers (Basel) 2022; 14:cancers14102348. [PMID: 35625953 PMCID: PMC9139326 DOI: 10.3390/cancers14102348] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/07/2022] [Accepted: 05/08/2022] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Recent findings in multiple myeloma have led to therapies which have improved patient life quality and expectancy. However, frequent relapse and drug resistance emphasize the need for more efficient therapeutic approaches. The discovery of non-coding RNAs as key actors in multiple myeloma has broadened the molecular landscape of this disease, together with classical epigenetic factors such as methylation and acetylation. microRNAs and long non-coding RNAs comprise the majority of the described non-coding RNAs dysregulated in multiple myeloma, while circular RNAs are recently emerging as promising molecular targets. This review provides a comprehensive overview of the most recent knowledge on this topic and suggests new therapeutic strategies. Abstract Despite advances in available treatments, multiple myeloma (MM) remains an incurable disease and represents a challenge in oncohematology. New insights into epigenetic factors contributing to MM development and progression have improved the knowledge surrounding its molecular basis. Beyond classical epigenetic factors, including methylation and acetylation, recent genome analyses have unveiled the importance of non-coding RNAs in MM pathogenesis. Non-coding RNAs have become of interest, as their dysregulation opens the door to new therapeutic approaches. The discovery, in the past years, of molecular techniques, such as CRISPR-Cas, has led to innovative therapies with potential benefits to achieve a better outcome for MM patients. This review summarizes the current knowledge on epigenetics and non-coding RNAs in MM pathogenesis.
Collapse
|
5
|
miRNA-seq and clinical evaluation in multiple myeloma: miR-181a overexpression predicts short-term disease progression and poor post-treatment outcome. Br J Cancer 2022; 126:79-90. [PMID: 34718359 PMCID: PMC8727627 DOI: 10.1038/s41416-021-01602-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/10/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Despite significant advances in multiple myeloma (MM) therapy, disease relapse and treatment resistance remain major obstacles in clinical management. Herein, we have studied the clinical utility of miRNAs in improving patients' risk-stratification and prognosis. METHODS miRNA-seq was performed in CD138+ plasma cells of MM, smoldering multiple myeloma (sMM) and monoclonal gammopathy of undetermined significance (MGUS) patients. The screening MM cohort consisted of 138 patients. miRNA levels of CD138+ plasma cells were quantified by RT-qPCR following 3'-end RNA polyadenylation. Disease progression and patients' death were used as clinical end-point events. Internal validation was conducted by bootstrap analysis. Clinical net benefit on disease prognosis was assessed by decision curve analysis. Kruykov et al. 2016 served as validation cohort (n = 151). RESULTS miRNA-seq highlighted miR-181a to be upregulated in MM vs. sMM/MGUS, and R-ISS III vs. I patients. Screening and validation cohorts confirmed the significantly higher risk for short-term progression and worse survival of the patients overexpressing miR-181a. Multivariate models integrating miR-181a with disease established markers led to superior risk-stratification and clinical benefit for MM prognosis. CONCLUSIONS CD138+ overexpression of miR-181a was strongly correlated with inferior disease outcome and contributed to superior prediction of MM patients early progression, supporting personalised prognosis and treatment decisions.
Collapse
|
6
|
Nano-Microparticle Platforms in Developing Next-Generation Vaccines. Vaccines (Basel) 2021; 9:vaccines9060606. [PMID: 34198865 PMCID: PMC8228777 DOI: 10.3390/vaccines9060606] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/03/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
The first vaccines ever made were based on live-attenuated or inactivated pathogens, either whole cells or fragments. Although these vaccines required the co-administration of antigens with adjuvants to induce a strong humoral response, they could only elicit a poor CD8+ T-cell response. In contrast, next-generation nano/microparticle-based vaccines offer several advantages over traditional ones because they can induce a more potent CD8+ T-cell response and, at the same time, are ideal carriers for proteins, adjuvants, and nucleic acids. The fact that these nanocarriers can be loaded with molecules able to modulate the immune response by inducing different effector functions and regulatory activities makes them ideal tools for inverse vaccination, whose goal is to shut down the immune response in autoimmune diseases. Poly (lactic-co-glycolic acid) (PLGA) and liposomes are biocompatible materials approved by the Food and Drug Administration (FDA) for clinical use and are, therefore, suitable for nanoparticle-based vaccines. Recently, another candidate platform for innovative vaccines based on extracellular vesicles (EVs) has been shown to efficiently co-deliver antigens and adjuvants. This review will discuss the potential use of PLGA-NPs, liposomes, and EVs as carriers of peptides, adjuvants, mRNA, and DNA for the development of next-generation vaccines against endemic and emerging viruses in light of the recent COVID-19 pandemic.
Collapse
|
7
|
Zhang Y, Zhang C, Wang J, Liu H, Wang M. Bone-Adipose Tissue Crosstalk: Role of Adipose Tissue Derived Extracellular Vesicles in Bone Diseases. J Cell Physiol 2021; 236:7874-7886. [PMID: 33993498 DOI: 10.1002/jcp.30414] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 04/08/2021] [Accepted: 04/26/2021] [Indexed: 11/08/2022]
Abstract
Bone is a metabolically active organ that undergoes constant remodeling throughout life. A failure of this process leads to pathological destructive bone diseases such as osteoporosis, rheumatoid arthritis, and osteoarthritis. Studies of the interplay between adipose tissue and bone system, have revealed that adipose tissue disorders (e.g. obesity) strongly influence the development of bone diseases. Adipokines secreted by adipose tissue play important roles in the crosstalk between bone and adipose tissue. Recently, extracellular vesicles (EVs) have been identified as a novel method of communication between different organs and have attracted increased attention in the field of bone remodeling process. Adipokines carried by EVs are known to play pivotal roles in bone remodeling processes including osteogenesis and osteoclastogenesis. In this review, we highlighted the role of adipose tissue derived EVs (EVs-AT) in the context of bone remodeling events and focused on the characteristics of EVs-AT and their components in the regulation of bone diseases. Moreover, we introduced the intriguing therapeutic application of EVs-AT in different pathological destructive bone diseases and proposed future directions for research on EVs-AT in bone diseases.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Oral and Maxillofacial Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, China.,Tianjin Clinical Research Center for Oral Diseases, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Cheng Zhang
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, China.,Tianjin Clinical Research Center for Oral Diseases, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China.,Department of Orthodontics, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Jiasheng Wang
- Department of Oral and Maxillofacial Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, China.,Tianjin Clinical Research Center for Oral Diseases, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Hao Liu
- Department of Oral and Maxillofacial Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, China.,Tianjin Clinical Research Center for Oral Diseases, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Muyao Wang
- Department of Oral and Maxillofacial Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, China.,Tianjin Clinical Research Center for Oral Diseases, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
8
|
Andalib A, Rashed S, Dehbashi M, Hajati J, Noorbakhsh F, Ganjalikhani-Hakemi M. The Upregulation of hsa-mir-181b-1 and Downregulation of Its Target CYLD in the Late-Stage of Tumor Progression of Breast Cancer. Indian J Clin Biochem 2020; 35:312-321. [PMID: 32647409 PMCID: PMC7326880 DOI: 10.1007/s12291-019-00826-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 03/11/2019] [Indexed: 02/05/2023]
Abstract
Some microRNAs are usually dysregulated in the cancers and influencing tumor behavior and progression. Hsa-miR-181b-1 and its target CYLD are involved in regulating the inflammatory pathways. This study aimed to investigate the expression levels of hsa-mir-181b-1 and CYLD in a cohort of breast tumor tissues and normal adjacent tissues to assess their association with breast cancer stages. A total number of 60 breast samples including cancerous and normal adjacent tissue specimens were collected. After pathological study, the expression of hsa-mir-181b-1 and CYLD were measured by qRT-PCR method. The hsa-mir-181b-1 expression level was significantly increased in breast tumor tissues compared to the controls. This increase was associated with the disease progression. Conversely, CYLD expression level was decreased in tumor samples compared to normal samples, significantly. ROC curve data added other prestigious information of hsa-mir-181b-1 and CYLD by defining cancer and healthy tissues with high specificity and sensitivity at a proposed cutoff point. Also, bioinformatic enrichment for the possible targets of mature sequence of "hsa-mir-181b-5p" was performed. Computational analysis showed the five most significant pathways including metabolic, cancer, calcium signaling, PI3K-Akt signaling and focal adhesion pathways which may be influenced by hsa-mir-181b-1. Thus, we suggested hsa-mir-181b-1 and CYLD might be involved in the pathogenesis of breast cancer and could be considered as two biomarkers for prediction, prognosis and diagnosis of the stages of the breast cancer.
Collapse
Affiliation(s)
- Alireza Andalib
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, 81746-73461 Iran
| | - Shadi Rashed
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, 81746-73461 Iran
| | - Moein Dehbashi
- Division of Genetics, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, 81746-73441 Iran
| | - Jamshid Hajati
- Department of Immunology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshid Noorbakhsh
- Department of Immunology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mazdak Ganjalikhani-Hakemi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, 81746-73461 Iran
| |
Collapse
|
9
|
Yan X, Gao M, Zhang P, Ouyang G, Mu Q, Xu K. MiR-181a functions as an oncogene by regulating CCND1 in multiple myeloma. Oncol Lett 2020; 20:758-764. [PMID: 32566002 PMCID: PMC7286114 DOI: 10.3892/ol.2020.11579] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 02/04/2020] [Indexed: 01/08/2023] Open
Abstract
MicroRNA-181a (miR-181a) has been demonstrated to be upregulated in patients with multiple myeloma (MM). In several studies, miR-181a has been demonstrated to be significantly overexpressed in MM; however, its potential role in development and progression of MM remains unknown. In the present study, the functions of miR-181a and the potential underlying molecular mechanisms in the pathogenesis of MM were examined. Increased expression of miR-181a was observed in bone marrow samples from patients with MM and the MM RPMI8226 cell line. The role of miR-181a was examined and it was demonstrated that it participated in the proliferation and migration processes of the MM cell line. Furthermore, it was demonstrated that the downregulation of miR-181a inhibited the expression of CCND1, a cell cycle regulatory gene, and caused cell cycle arrest in MM cells. The results of the present study suggested that miR-181a functions as an onco-miRNA in MM, which serves regulatory roles by upregulating expression of CCND1 and may therefore serve as a potential target in patients with MM.
Collapse
Affiliation(s)
- Xiao Yan
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Minjie Gao
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Ping Zhang
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Guifang Ouyang
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Qitian Mu
- Stem Cell Laboratory, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Kaihong Xu
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
10
|
Braicu C, Gulei D, Raduly L, Harangus A, Rusu A, Berindan-Neagoe I. Altered expression of miR-181 affects cell fate and targets drug resistance-related mechanisms. Mol Aspects Med 2019; 70:90-105. [PMID: 31703947 DOI: 10.1016/j.mam.2019.10.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/22/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNAs) are non-coding transcripts which regulate genetic and epigenetic events by interfering with mRNA translation. miRNAs are involved in regulation of cell fate due to their ability of interfering with physiological or pathological processes. In this review paper, we evaluate the role of miR-181 family members as prognostic or diagnostic markers or therapeutic targets in malignant pathologies in connection with the main hallmarks of cancer that are modulated by the family. Also, we take over the dual role of this family in dependency with the tumour suppressor and oncogenic features presented in cell and cancer type specific manner. Restoration of the altered expression levels contributes to the activation of cell death pathways or to a reduction in the invasion and migration mechanism; moreover, the mechanism of drug resistance is also modulated by miR-181 sequences with important applications in therapeutic strategies for malignant cells sensitisation. Overall, the main miR-181 family regulatory mechanisms are presented in a cancer specific context, emphasizing the possible clinical application of this family in terms of novel diagnosis and therapy approaches.
Collapse
Affiliation(s)
- Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| | - Diana Gulei
- MedFuture Research Center for Advanced Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lajos Raduly
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Antonia Harangus
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania; "Leon Daniello" Pneumophtisiology Clinic, 6 Bogdan Petriceicu Hasdeu Street, 400332, Cluj-Napoca, Romania.
| | | | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania; MedFuture Research Center for Advanced Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania; Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania.
| |
Collapse
|
11
|
Yang H, Li Y, Peng Z, Wang Y. Overexpression of miR-20a promotes the progression of osteosarcoma by directly targeting QKI2. Oncol Lett 2019; 18:87-94. [PMID: 31289476 PMCID: PMC6540454 DOI: 10.3892/ol.2019.10313] [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: 01/31/2018] [Accepted: 01/22/2019] [Indexed: 11/15/2022] Open
Abstract
Osteosarcoma (OS) is the most common type of malignant primary bone neoplasm. Although the application of neoadjuvant chemotherapy has improved the 5-year survival rate of patients suffering from OS, prognosis remains poor. Therefore, it is important to elucidate the molecular mechanisms underlying the occurrence, progression and metastasis of OS. The RNA-binding protein Quaking (QKI) is a member of the STAR family of proteins, and can function as a tumor suppressor gene to suppress the occurrence and progression of a variety of tumors; however, the role of QKI in OS remains to be fully elucidated. In the present study, it was identified that the expression of QKI2 was downregulated in OS using western blot analysis. In addition, subsequent functional investigations, including MTT, Transwell invasion and migration assays, revealed that QKI2 inhibited the proliferation, invasion and migration of an OS cell line in vitro. By implementing a series of experimental techniques in molecular biology, including reverse transcription-quantitative polymerase chain reaction and a double fluorescence reporter assay, it was demonstrated that the expression of miR-20a was high and inhibited the expression of QKI2 in OS. In conclusion, it was revealed that aberrantly upregulated miR-20a inhibited the expression of QKI2 in OS by targeting QKI2 mRNA, subsequently promoting the proliferation, migration and invasion of OS cells.
Collapse
Affiliation(s)
- Hongbo Yang
- Department of Orthopedic Surgery, Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia 024000, P.R. China
| | - Yongli Li
- Department of Tumor Radiotherapy, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150000, P.R. China
| | - Zhibin Peng
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yansong Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
12
|
Abstract
This study aims to investigate the role of miR-181a in multiple myeloma (MM). Fresh peripheral blood and bone marrows were collected. Expression of miR-181a, BCL-2 mRNA, and NOVA1 mRNA was detected by RT-qPCR. The correlation between miR-181a and clinical features of MM was further analyzed. miR-181a in serum and bone marrow mononuclear cells of MM patients were significantly higher. And, miR-181a level was significantly higher in MM Durie-Salmon stage III than that in stage I+II. miR-181a was positively correlated to Durie-Salmon staging, age, kidney injury, bone injury, β2-MG whereas negatively related to red blood cell, hemoglobin, and albumin. Additionally, BCL-2 and NOVA1 were predicted to be downstream targets of miR-181a. BCL-2 mRNA was significantly higher in the bone marrow mononuclear cells from MM patients. To sum up, the miR-181a expression is increased in peripheral blood and bone marrow of MM patients and is closely related to the clinical pathological indicators of MM.
Collapse
Affiliation(s)
- Ruili Yuan
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an
| | - Ni Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an
| | - Jinyu Yang
- Department of Clinical Laboratory, An’kang Hospital of Traditional Chinese Medicine, An’kang
| | - Jing Peng
- Department of Clinical Laboratory, Xi’an Hospital of Traditional Chinese Medicine, Xi’an, China
| | - Lina Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an
| | - Xuan Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an
| |
Collapse
|
13
|
Zhu B, Ju S, Chu H, Shen X, Zhang Y, Luo X, Cong H. The potential function of microRNAs as biomarkers and therapeutic targets in multiple myeloma. Oncol Lett 2018; 15:6094-6106. [PMID: 29731841 PMCID: PMC5920744 DOI: 10.3892/ol.2018.8157] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/10/2018] [Indexed: 12/30/2022] Open
Abstract
Multiple myeloma (MM), accounting for ~1% of all types of human cancer and 13% of all hematological malignancies, is characterized by the malignant proliferation of monoclonal plasma cells (PCs) in the bone marrow. MM leads to end stage organ impairment, including bone lesions, renal dysfunction, hypercalcemia and anemia. So far, the specific pathogenesis of MM remains unclear and no early-stage sensitive biomarker of MM has been well characterized. Furthermore, treating MM is difficult, as the majority of patients eventually relapse or become refractory following treatment using presently available methods. To date, a number of studies have demonstrated that microRNAs (miRNAs) may serve crucial functions in the progression of numerous cancers, including MM. During the tumorigenesis and pathogenesis of MM, there are multiple carcinogenic events that involve the pernicious transformation from normal to malignant PCs. miRNAs, as oncogenes or tumor suppressors, regulate MM progression-related signaling pathways. In the present review, the up-to-date preliminary basic studies and associated clinical works on the underlying mechanisms of aberrant miRNA profiling in MM have been summarized, including an evaluation of its value as a potential biomarker and a novel therapeutic strategy for MM.
Collapse
Affiliation(s)
- Bingying Zhu
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Shaoqing Ju
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Haidan Chu
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Xianjuan Shen
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Yan Zhang
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Xi Luo
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Hui Cong
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| |
Collapse
|
14
|
Jiang Y, Chang H, Chen G. Effects of microRNA-20a on the proliferation, migration and apoptosis of multiple myeloma via the PTEN/PI3K/AKT signaling pathway. Oncol Lett 2018; 15:10001-10007. [PMID: 29963125 DOI: 10.3892/ol.2018.8555] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 01/03/2018] [Indexed: 12/13/2022] Open
Abstract
Multiple myeloma (MM) is a heterogeneous disease with a poor prognosis. Circulating microRNAs (miRNAs) have shown potential as non-invasive prognostic biomarkers for heterogeneous diseases. miR-20a has been shown involved in various human cancers, and the phosphatase and tensin homolog/phosphoinositide 3-kinase/protein kinase B (PTEN/P13K/Akt) signaling pathway plays a key role in cell proliferation, migration and apoptosis. Here, we investigated the effect of miR-20a on the PTEN/PI3K/Akt signaling pathway during MM cell proliferation, migration and apoptosis. Reverse transcription quantitative polymerase chain reaction was applied to detect miR-20a expression in plasma from 30 MM patients and MM cell lines. CCK-8 assays, Transwell assays, Annexin V/PI double-staining and western blotting were performed to examine the protein expressions of PTEN, PI3K and Akt during cellullar proliferation, migration, cycling, and apoptosis. Significant upregulation of miR-20a and deregulation of PTEN were observed in MM cells. We also identified PTEN as a downstream target gene of miR-20a, which bound to the 3'-untranslated region of PTEN. Overexpression of miR-20a was associated with decreased PTEN expression, and treatment with miR-20a inhibitors decreased cell proliferation, migration and clonogenicity and reduced the protein expressions of PI3K and p-Akt but increased PTEN protein expression compared with blank and negative control groups. Taken together, these results showed that inhibition of miR-20a suppresses MM progression by modulating the PTEN/PI3K/Akt signaling pathway. These findings suggest that miR-20a may be a novel molecular therapeutic target for the treatment of MM.
Collapse
Affiliation(s)
- Yanxia Jiang
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Hong Chang
- Division of Molecular and Cellular Biology, Toronto General Research Institute, Toronto, ON M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Guoan Chen
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| |
Collapse
|
15
|
Ba Z, Gu L, Hao S, Wang X, Cheng Z, Nie G. Downregulation of lncRNA CASC2 facilitates osteosarcoma growth and invasion through miR-181a. Cell Prolif 2017; 51. [PMID: 29194827 DOI: 10.1111/cpr.12409] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 10/16/2017] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Long non-coding RNA cancer susceptibility candidate 2 (CASC2) is a novel lncRNA and has been indicated as playing tumour suppressor gene in several tumours. However, the role of CASC2 in osteosarcoma is still uncovered. MATERIALS AND METHODS The CASC2 and miR-181a expressions were measured via qRT-PCR. CCK-8 assay and colony formation assay were performed to determine the cell growth, and transwell assay was performed to assess the cell invasion. RESULTS We showed that CASC2 expression was downregulated in osteosarcoma samples and cell lines. Moreover, we showed that downregulated expression of CASC2 was correlated with advanced TNM stage. Furthermore, overexpression of CASC2 inhibited osteosarcoma cell proliferation, colony formation, and invasion. In addition, we indicated that ectopic expression of CASC2 suppressed miR-181a expression and enhanced the expression of Ras association domain family member 6 (RASSF6), PTEN and ATM in osteosarcoma cell, which were the direct target gene of miR-181a. Moreover, we indicated that RASSF6 expression was downregulated in osteosarcoma samples and cell lines and downregulated expression of RASSF6 was correlated with advanced TNM stage. We found that the expression of RASSF6 was positively correlated with the expression of CASC2 in osteosarcoma tissues. Ectopic expression of CASC2 suppressed the osteosarcoma cell proliferation, colony formation and invasion through regulating RASSF6 expression. CONCLUSIONS Our data illuminated that CASC2 acted as a tumour suppressor in osteosarcoma progression.
Collapse
Affiliation(s)
- Zhiwen Ba
- Department of Orthopedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, 150040, China
| | - Lili Gu
- Department of Orthopedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, 150040, China
| | - Songnan Hao
- Department of Orthopedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, 150040, China
| | - Xiaofang Wang
- Department of Infectious Disease, The Forth Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, China
| | - Zhenping Cheng
- Department of Orthopedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, 150040, China
| | - Guangchen Nie
- Department of Orthopedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, 150040, China
| |
Collapse
|
16
|
Zhang X, Song H, Qiao S, Liu J, Xing T, Yan X, Li H, Wang N. MiR-17-5p and miR-20a promote chicken cell proliferation at least in part by upregulation of c-Myc via MAP3K2 targeting. Sci Rep 2017; 7:15852. [PMID: 29158522 PMCID: PMC5696470 DOI: 10.1038/s41598-017-15626-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/30/2017] [Indexed: 12/14/2022] Open
Abstract
The miR-17-92 cluster has been well studied in mammals but less extensively studied in birds. Here, we demonstrated that miR-17-92 cluster overexpression promoted the proliferation of DF1 cells and immortalized chicken preadipocytes (ICPA-1), and miR-17-5p and miR-20a, members of the miR-17-92 cluster, targeted MAP3K2. Further analysis showed that MAP3K2 overexpression reduced the proliferation of DF1 and ICPA-1 cells and attenuated the promotive effect of the miR-17-92 cluster on cell proliferation. Downstream gene expression analysis of the MAPK signalling pathway showed that MAP3K2 overexpression decreased c-Myc expression; in contrast, MAP3K2 knockdown using RNA interference and miR-17-92 cluster overexpression increased c-Myc expression. Furthermore, c-Myc overexpression promoted miR-17-92 cluster expression and DF1 cell proliferation. Taken together, these data indicated that miR-17-92 promotes chicken cell proliferation at least in part by the upregulation of c-Myc via targeting MAP3K2, and the miR-17-92 cluster, c-Myc and E2F1 form a complex regulatory network in chicken cell proliferation.
Collapse
Affiliation(s)
- Xiaofei Zhang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin, 150030, Heilongjiang, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, Heilongjiang, China.,Key Laboratory of Animal Cells and Genetic Engineering of Heilongjiang Province, Harbin, 150030, Heilongjiang, China
| | - He Song
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin, 150030, Heilongjiang, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, Heilongjiang, China.,Key Laboratory of Animal Cells and Genetic Engineering of Heilongjiang Province, Harbin, 150030, Heilongjiang, China
| | - Shupei Qiao
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin, 150030, Heilongjiang, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, Heilongjiang, China.,Key Laboratory of Animal Cells and Genetic Engineering of Heilongjiang Province, Harbin, 150030, Heilongjiang, China
| | - Jing Liu
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin, 150030, Heilongjiang, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, Heilongjiang, China.,Key Laboratory of Animal Cells and Genetic Engineering of Heilongjiang Province, Harbin, 150030, Heilongjiang, China
| | - Tianyu Xing
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin, 150030, Heilongjiang, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, Heilongjiang, China.,Key Laboratory of Animal Cells and Genetic Engineering of Heilongjiang Province, Harbin, 150030, Heilongjiang, China
| | - Xiaohong Yan
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin, 150030, Heilongjiang, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, Heilongjiang, China.,Key Laboratory of Animal Cells and Genetic Engineering of Heilongjiang Province, Harbin, 150030, Heilongjiang, China
| | - Hui Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin, 150030, Heilongjiang, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, Heilongjiang, China.,Key Laboratory of Animal Cells and Genetic Engineering of Heilongjiang Province, Harbin, 150030, Heilongjiang, China
| | - Ning Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin, 150030, Heilongjiang, China. .,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, 150030, Heilongjiang, China. .,Key Laboratory of Animal Cells and Genetic Engineering of Heilongjiang Province, Harbin, 150030, Heilongjiang, China.
| |
Collapse
|
17
|
Lei Z, Ma X, Li H, Zhang Y, Gao Y, Fan Y, Li X, Chen L, Xie Y, Chen J, Wu S, Tang L, Zhang X. Up-regulation of miR-181a in clear cell renal cell carcinoma is associated with lower KLF6 expression, enhanced cell proliferation, accelerated cell cycle transition, and diminished apoptosis. Urol Oncol 2017; 36:93.e23-93.e37. [PMID: 29066014 DOI: 10.1016/j.urolonc.2017.09.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 09/14/2017] [Accepted: 09/18/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Dysregulated expression of miR-181a accompanies tumorigenesis in many human cancers. However, in clear cell renal cell carcinoma (ccRCC), the role of miR-181a remains unclear. The aim of this study was to investigate biological functions of miR-181a and its expression levels in ccRCC tissues and cancer cell lines. MATERIAL AND METHODS Expression levels of miR-181a in samples of ccRCC tumors and adjacent nontumor tissues from 42 patients as well as in 786-O, 769-P, A498, and CAKI-1 ccRCC cell lines were determined by quantitative real-time polymerase chain reaction. Potential targets of miR-181a were predicted using bioinformatic approaches and then verified by using the luciferase reporter assay. The effects of miR-181a on cell proliferation, colony formation, cell cycle progression, and apoptosis were investigated in ccRCC cell lines transfected with specific miR-181a mimic and inhibitor. RESULTS We found that miR-181a expression was up-regulated in ccRCC tissues and cell lines. The expression level of miR-181a significantly correlated with the tumor size, tumor/node/metastasis staging, and Fuhrman grade. Luciferase assays showed that KLF6 was a target of miR-181a. KLF6 expression was inversely correlated with the level of miR-181a. Overexpression of miR-181a led to reduced KLF6 mRNA and protein levels, whereas mutations of the potential miR-181a binding sites in the KLF6 gene abrogated this inhibitory effect. Furthermore, overexpression of miR-181a promoted proliferation and G1/S cell cycle transition, as well as inhibited apoptosis by down-regulating KLF6 in ccRCC cells. CONCLUSIONS miR-181a is up-regulated in ccRCC and may act as a tumor promoting factor by targeting KLF6 expression. Manipulating miR-181a may provide a beneficial effect in the treatment of ccRCC.
Collapse
Affiliation(s)
- Zhenwei Lei
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Xin Ma
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Hongzhao Li
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Yu Zhang
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Yu Gao
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Yang Fan
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Xintao Li
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Luyao Chen
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Yongpeng Xie
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Jianwen Chen
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Shengpan Wu
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Lu Tang
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Xu Zhang
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army Medical School, Chinese People's Liberation Army General Hospital, Beijing, PR China.
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Multiple myeloma remains an incurable disease, largely due to the tumor-supportive role of the bone marrow microenvironment. Bone marrow adipose tissue (BMAT) is one component of the fertile microenvironment which is believed to contribute to myeloma progression and drug resistance, as well as participate in a vicious cycle of osteolysis and tumor growth. RECENT FINDINGS MicroRNAs (miRNAs) have recently emerged as instrumental regulators of cellular processes that enable the development and dissemination of cancer. This review highlights the intersection between two emerging research fields and pursues the scientific and clinical implications of miRNA transfer between BMAT and myeloma cells. This review provides a concise and provocative summary of the evidence to support exosome-mediated transfer of tumor-supportive miRNAs. The work may prompt researchers to better elucidate the mechanisms by which this novel means of genetic communication between tumor cells and their environment could someday yield targeted therapeutics.
Collapse
Affiliation(s)
- Luna Soley
- Maine Medical Center Research Institute, Scarborough, ME, 04074, USA
| | - Carolyne Falank
- Maine Medical Center Research Institute, Scarborough, ME, 04074, USA
| | - Michaela R Reagan
- Maine Medical Center Research Institute, Scarborough, ME, 04074, USA.
- University of Maine, Orono, ME, 04469, USA.
- Sackler School of Graduate Biomedical Sciences and School of Medicine, Tufts University, Boston, MA, 02111, USA.
| |
Collapse
|
19
|
Shen X, Ye Y, Qi J, Shi W, Wu X, Ni H, Cong H, Ju S. Identification of a novel microRNA, miR-4449, as a potential blood based marker in multiple myeloma. Clin Chem Lab Med 2017; 55:748-754. [PMID: 27155004 DOI: 10.1515/cclm-2015-1108] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 04/04/2016] [Indexed: 01/07/2023]
Abstract
BACKGROUND miRNAs act in diverse biological processes including development, cell growth, apoptosis, and hematopoiesis, suggesting their role in cancer. METHODS We examined the miRNAs perturbed in CD138+ primary multiple myeloma (MM) cells, using microarray analysis and real-time quantitative PCR (RT-qPCR). Serum miR-4449 expression levels were detected from 71 primary MM patients and 46 healthy controls by RT-qPCR. RESULTS Our analysis revealed up-regulation of 54 and down-regulation of 28 miRNAs in MM subjects compared to healthy controls. miR-4449 has not been reported in MM. It was found that the relative expression of bone marrow miR-4449 in MM patients (2.14±1.42) was higher than that in healthy controls (0.815±0.165) (U=8, p=0.0093). The relative expression of serum miR-4449 in MM patients (2.11±2.10) was significantly higher than that in healthy controls (0.357±0.235) (U=374, p<0.0001) and was significantly correlated with β2M, λ light and κ light chain concentration (r=0.480, p=0.0003; r=0.560, p<0.0001; r=0.560, p<0.0001), but not correlated with the lactate dehydrogenase (LDH) concentration (r=0.247, p=0.0611). The area under the curve (AUC) of the receiver-operating characteristics (ROC) curve of serum miR-4449 was 0.885 (95% CI, 0.826-0.945), which is higher than for other markers. Combining miR-4449, λ light chain, and β2M together, the sensitivity was highest compared with λ light chain or β2M alone, or combined. CONCLUSIONS The expression levels of serum miR-4449 in MM patients were significantly higher than in healthy controls, suggesting that it may prove to be useful in the auxiliary diagnosis of MM.
Collapse
Affiliation(s)
- Xianjuan Shen
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, P.R
| | - Yan Ye
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, P.R
| | - Jing Qi
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, P.R
| | - Wei Shi
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, P.R
| | - Xinhua Wu
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, P.R
| | - Hongbing Ni
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, P.R
| | - Hui Cong
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, P.R
| | - Shaoqing Ju
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, JS, P.R
| |
Collapse
|
20
|
Zhang X, Chen Y, Zhao P, Zang L, Zhang Z, Wang X. MicroRNA-19a functions as an oncogene by regulating PTEN/AKT/pAKT pathway in myeloma. Leuk Lymphoma 2016; 58:932-940. [PMID: 27830963 DOI: 10.1080/10428194.2016.1213827] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
MicroRNAs (miRNAs) are involved in human cancers including myeloma. MiR-19a is one of the oncogenic miR-17-92 clusters, which is identified as a key oncogenic component in many cancers. Nevertheless, the function of miR-19a in myeloma has not been totally elucidated. The purpose of this study is to investigate the biological functions of miR-19a in MM. In vitro, we detected that the miR-19a-3p is overexpressed in myeloma cells. The proliferation and invision of myeloma cells are analyzed by MTT and BD matrigel assays, respectively. Western blot was performed to evaluate the expression of apoptotic/drug resistance-related main control proteins BCL-2 and MDR1 in myeloma cells after transfected with miR-19a-3p. Finally, we found miR-19a acts as an oncogene in MM by promoting cell proliferation/invision and inhibiting apoptosis. Additionally, We further showed that the mRNA and protein of BCL-2 and MDR were upregulated significantly after elevated expression of miR-19a, the process of which was regulated by PTEN/AKT/pAKT-signaling pathway. Our results suggest that miR-19a acted as an oncogenic miRNA by targeting PTEN in myeloma. This novel miR-19a/PTEN/AKT axis sheds new light on the mechanisms underlying apoptosis and invision and may provide potentially therapeutic targets for the treatment of myeloma.
Collapse
Affiliation(s)
- Xiaoying Zhang
- a Department of Hematology, Key Laboratory of Cancer Prevention and Therapy , Tianjin Medical University, Cancer Hospital of Tianjin , Tianjin , China
| | - Yafang Chen
- a Department of Hematology, Key Laboratory of Cancer Prevention and Therapy , Tianjin Medical University, Cancer Hospital of Tianjin , Tianjin , China
| | - Pan Zhao
- a Department of Hematology, Key Laboratory of Cancer Prevention and Therapy , Tianjin Medical University, Cancer Hospital of Tianjin , Tianjin , China
| | - Li Zang
- a Department of Hematology, Key Laboratory of Cancer Prevention and Therapy , Tianjin Medical University, Cancer Hospital of Tianjin , Tianjin , China
| | - Zhiqing Zhang
- b Department of Neurology , The Fourth Central Hospital , Tianjin , China
| | - Xiaofang Wang
- a Department of Hematology, Key Laboratory of Cancer Prevention and Therapy , Tianjin Medical University, Cancer Hospital of Tianjin , Tianjin , China
| |
Collapse
|