1
|
Ban GI, Puviindran V, Xiang Y, Nadesan P, Tang J, Ou J, Guardino N, Nakagawa M, Browne M, Wallace A, Ishikawa K, Shimada E, Martin JT, Diao Y, Kirsch DG, Alman BA. The COMPASS complex maintains the metastatic capacity imparted by a subpopulation of cells in UPS. iScience 2024; 27:110187. [PMID: 38989451 PMCID: PMC11233968 DOI: 10.1016/j.isci.2024.110187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 04/20/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024] Open
Abstract
Intratumoral heterogeneity is common in cancer, particularly in sarcomas like undifferentiated pleomorphic sarcoma (UPS), where individual cells demonstrate a high degree of cytogenic diversity. Previous studies showed that a small subset of cells within UPS, known as the metastatic clone (MC), as responsible for metastasis. Using a CRISPR-based genomic screen in-vivo, we identified the COMPASS complex member Setd1a as a key regulator maintaining the metastatic phenotype of the MC in murine UPS. Depletion of Setd1a inhibited metastasis development in the MC. Transcriptome and chromatin sequencing revealed COMPASS complex target genes in UPS, such as Cxcl10, downregulated in the MC. Deleting Cxcl10 in non-MC cells increased their metastatic potential. Treating mice with human UPS xenografts with a COMPASS complex inhibitor suppressed metastasis without affecting tumor growth in the primary tumor. Our data identified an epigenetic program in a subpopulation of sarcoma cells that maintains metastatic potential.
Collapse
Affiliation(s)
- Ga I. Ban
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Vijitha Puviindran
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Yu Xiang
- Department of Cell Biology and Duke Regeneration Center, Duke University School of Medicine, Durham, NC, USA
| | - Puvi Nadesan
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Jackie Tang
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Jianhong Ou
- Department of Cell Biology and Duke Regeneration Center, Duke University School of Medicine, Durham, NC, USA
| | - Nicholas Guardino
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Makoto Nakagawa
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - MaKenna Browne
- Department of Cell Biology and Duke Regeneration Center, Duke University School of Medicine, Durham, NC, USA
| | - Asjah Wallace
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Koji Ishikawa
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Eijiro Shimada
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - John T. Martin
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Yarui Diao
- Department of Cell Biology and Duke Regeneration Center, Duke University School of Medicine, Durham, NC, USA
| | - David G. Kirsch
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
- The Princes Margaret Cancer Centre, Department of Radiation Oncology, University Health Network and the University of Toronto, Toronto, ON, Canada
| | - Benjamin A. Alman
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
2
|
Zheng Y, Sukocheva O, Tse E, Neganova M, Aleksandrova Y, Zhao R, Chubarev V, Fan R, Liu J. MicroRNA-183 cluster: a promising biomarker and therapeutic target in gastrointestinal malignancies. Am J Cancer Res 2023; 13:6147-6175. [PMID: 38187051 PMCID: PMC10767355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/29/2023] [Indexed: 01/09/2024] Open
Abstract
Small non-coding RNAs (microRNA, miR), powerful epigenetic regulators, were found involved in the regulation of most biological functions via post-translational inhibition of protein expression. Increased expression of pro-oncogenic miRs (known as miR cancer biomarkers) and inhibition of pro-apoptotic miR expression have been demonstrated in different tumors. The recently identified miR-183 was found implicated in gastrointestinal tumor metabolism regulation. Elevated miR-183 expression and cancer-promoting effects were reported in esophageal and colorectal cancers, which was partially contradicted by controversial data observed in gastric cancers. Anti-cancer effect of miR-183 in gastric cancer cells was associated with the Bim-1 and Ezrin genes regulation. Many studies indicated that miR-183 can inhibit tumor suppressor genes in most cell lines, promoting tumor cell proliferation and migration. Increased miR-183 level results in the downregulation of FOXO1, PDCD4, and other tumor suppressor genes in gastrointestinal tumor cells. MiR-183 also influences the signaling of PI3K/AKT/mTOR, Wnt/β-catenin, and Bcl-2/p53 signaling pathways. Mir-183 inhibits apoptosis and autophagy, and promotes epithelial-to-mesenchymal transition, cancer cell proliferation, and migration. Accordingly, gastrointestinal cancer occurrence, development of chemoradiotherapy resistance, recurrence/metastasis, and prognosis were associated with miR-183 expression. The current study assessed reported miR-183 functions and signaling, providing new insights for the diagnosis and treatment of gastrointestinal malignancies.
Collapse
Affiliation(s)
- Yufei Zheng
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou UniversityZhengzhou 450052, Henan, China
| | - Olga Sukocheva
- Department of Hepatology, Royal Adelaide HospitalAdelaide, SA 5000, Australia
| | - Edmund Tse
- Department of Hepatology, Royal Adelaide HospitalAdelaide, SA 5000, Australia
| | - Margarita Neganova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of SciencesSevernij Pr. 1, Chernogolovka 142432, Russia
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of SciencesAkad. Arbuzov St. 8, Kazan 420088, Russia
| | - Yulia Aleksandrova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of SciencesSevernij Pr. 1, Chernogolovka 142432, Russia
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of SciencesAkad. Arbuzov St. 8, Kazan 420088, Russia
| | - Ruiwen Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou UniversityZhengzhou 450052, Henan, China
| | - Vladimir Chubarev
- Sechenov First Moscow State Medical University (Sechenov University)8-2 Trubetskaya St., Moscow 119991, Russia
| | - Ruitai Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou UniversityZhengzhou 450052, Henan, China
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou UniversityZhengzhou 450052, Henan, China
| |
Collapse
|
3
|
Xiao Y, Hu Y, Liu S. Non-coding RNAs: a promising target for early metastasis intervention. Chin Med J (Engl) 2023; 136:2538-2550. [PMID: 37442775 PMCID: PMC10617820 DOI: 10.1097/cm9.0000000000002619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Indexed: 07/15/2023] Open
Abstract
ABSTRACT Metastases account for the overwhelming majority of cancer-associated deaths. The dissemination of cancer cells from the primary tumor to distant organs involves a complex process known as the invasion-metastasis cascade. The underlying biological mechanisms of metastasis, however, remain largely elusive. Recently, the discovery and characterization of non-coding RNAs (ncRNAs) have revealed the diversity of their regulatory roles, especially as key contributors throughout the metastatic cascade. Here, we review recent progress in how three major types of ncRNAs (microRNAs, long non-coding RNAs, and circular RNAs) are involved in the multistep procedure of metastasis. We further examine interactions among the three ncRNAs as well as current progress in their regulatory mechanisms. We also propose the prevention of metastasis in the early stages of cancer progression and discuss current translational studies using ncRNAs as targets for metastasis diagnosis and treatments. These studies provide insights into developing more effective strategies to target metastatic relapse.
Collapse
Affiliation(s)
- Yi Xiao
- Department of Stomatology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yijun Hu
- Clinical Research Center, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Shanrong Liu
- Department of Laboratory Diagnostics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
4
|
Kaur J, Saul D, Doolittle ML, Farr JN, Khosla S, Monroe DG. MicroRNA- 19a- 3p Decreases with Age in Mice and Humans and Inhibits Osteoblast Senescence. JBMR Plus 2023; 7:e10745. [PMID: 37283656 PMCID: PMC10241091 DOI: 10.1002/jbm4.10745] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 06/08/2023] Open
Abstract
Aging is a major risk factor for most chronic diseases, including osteoporosis, and is characterized by an accumulation of senescent cells in various tissues. MicroRNAs (miRNAs) are critical regulators of bone aging and cellular senescence. Here, we report that miR-19a-3p decreases with age in bone samples from mice as well as in posterior iliac crest bone biopsies of younger versus older healthy women. miR-19a-3p also decreased in mouse bone marrow stromal cells following induction of senescence using etoposide, H2O2, or serial passaging. To explore the transcriptomic effects of miR-19a-3p, we performed RNA sequencing of mouse calvarial osteoblasts transfected with control or miR-19a-3p mimics and found that miR-19a-3p overexpression significantly altered the expression of various senescence, senescence-associated secretory phenotype-related, and proliferation genes. Specifically, miR-19a-3p overexpression in nonsenescent osteoblasts significantly suppressed p16 Ink4a and p21 Cip1 gene expression and increased their proliferative capacity. Finally, we established a novel senotherapeutic role for this miRNA by treating miR-19a-3p expressing cells with H2O2 to induce senescence. Interestingly, these cells exhibited lower p16 Ink4a and p21 Cip1 expression, increased proliferation-related gene expression, and reduced SA-β-Gal+ cells. Our results thus establish that miR-19a-3p is a senescence-associated miRNA that decreases with age in mouse and human bones and is a potential senotherapeutic target for age-related bone loss. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Japneet Kaur
- Division of Endocrinology, Department of MedicineMayo Clinic College of MedicineRochesterMNUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
| | - Dominik Saul
- Division of Endocrinology, Department of MedicineMayo Clinic College of MedicineRochesterMNUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
| | - Madison L. Doolittle
- Division of Endocrinology, Department of MedicineMayo Clinic College of MedicineRochesterMNUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
| | - Joshua N. Farr
- Division of Endocrinology, Department of MedicineMayo Clinic College of MedicineRochesterMNUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
| | - Sundeep Khosla
- Division of Endocrinology, Department of MedicineMayo Clinic College of MedicineRochesterMNUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
| | - David G. Monroe
- Division of Endocrinology, Department of MedicineMayo Clinic College of MedicineRochesterMNUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
| |
Collapse
|
5
|
Hebert JD, Neal JW, Winslow MM. Dissecting metastasis using preclinical models and methods. Nat Rev Cancer 2023; 23:391-407. [PMID: 37138029 DOI: 10.1038/s41568-023-00568-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/27/2023] [Indexed: 05/05/2023]
Abstract
Metastasis has long been understood to lead to the overwhelming majority of cancer-related deaths. However, our understanding of the metastatic process, and thus our ability to prevent or eliminate metastases, remains frustratingly limited. This is largely due to the complexity of metastasis, which is a multistep process that likely differs across cancer types and is greatly influenced by many aspects of the in vivo microenvironment. In this Review, we discuss the key variables to consider when designing assays to study metastasis: which source of metastatic cancer cells to use and where to introduce them into mice to address different questions of metastasis biology. We also examine methods that are being used to interrogate specific steps of the metastatic cascade in mouse models, as well as emerging techniques that may shed new light on previously inscrutable aspects of metastasis. Finally, we explore approaches for developing and using anti-metastatic therapies, and how mouse models can be used to test them.
Collapse
Affiliation(s)
- Jess D Hebert
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Joel W Neal
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Monte M Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
6
|
Lee EY, Kim YH, Rayhan MA, Kang HG, Kim JH, Park JW, Park SY, Lee SH, You HJ. New established cell lines from undifferentiated pleomorphic sarcoma for in vivo study. BMB Rep 2023; 56:258-264. [PMID: 36789562 PMCID: PMC10140480 DOI: 10.5483/bmbrep.2022-0209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 11/06/2023] Open
Abstract
As a high-grade soft-tissue sarcoma (STS), undifferentiated pleomorphic sarcoma (UPS) is highly recurrent and malignant. UPS is categorized as a tumor of uncertain differentiation and has few options for treatment due to its lack of targetable genetic alterations. There are also few cell lines that provide a representative model for UPS, leading to a dearth of experimental research. Here, we established and characterized new cell lines derived from two recurrent UPS tissues. Cells were obtained from UPS tissues by mincing, followed by extraction or dissociation using enzymes and culture in a standard culture environment. Cells were maintained for several months without artificial treatment, and some cell clones were found to be tumorigenic in an immunodeficient mouse model. Interestingly, some cells formed tumors in vivo when injected after aggregation in a non-adherent culture system for 24 h. The tissues from in vivo study and tissues from patients shared common histological characteristics. Pathways related to the cell cycle, such as DNA replication, were enriched in both cell clones. Pathways related to cell-cell adhesion and cell-cell signaling were also enriched, suggesting a role of the mesenchymal-to-epithelial transition for tumorigenicity in vivo. These new UPS cell lines may facilitate research to identify therapeutic strategies for UPS. [BMB Reports 2023; 56(4): 258-264].
Collapse
Affiliation(s)
- Eun-Young Lee
- Cancer Microenvironment Branch, Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Young-Ho Kim
- Diagnostics and Therapeutics Technology Branch, Division of Technology Convergence, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Md Abu Rayhan
- Cancer Microenvironment Branch, Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Hyun Guy Kang
- Department of Cancer Biomedical Science, NCC-GCSP, National Cancer Center, Goyang 10408, Korea
- Department of Orthopaedic Surgery, Hospital, National Cancer Center, Goyang 10408, Korea
| | - June Hyuk Kim
- Department of Cancer Biomedical Science, NCC-GCSP, National Cancer Center, Goyang 10408, Korea
- Department of Orthopaedic Surgery, Hospital, National Cancer Center, Goyang 10408, Korea
| | - Jong Woong Park
- Department of Orthopaedic Surgery, Hospital, National Cancer Center, Goyang 10408, Korea
- Department of Cancer Control and Population Health, NCC-GCSP, National Cancer Center, Goyang 10408, Korea
| | - Seog-Yun Park
- Department of Pathology, National Cancer Center Hospital, Goyang 10408, Korea
| | - So Hee Lee
- Cancer Microenvironment Branch, Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Hye Jin You
- Cancer Microenvironment Branch, Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Korea
- Department of Cancer Biomedical Science, NCC-GCSP, National Cancer Center, Goyang 10408, Korea
| |
Collapse
|
7
|
Patel R, Mowery YM, Qi Y, Bassil AM, Holbrook M, Xu ES, Hong CS, Himes JE, Williams NT, Everitt J, Ma Y, Luo L, Selitsky SR, Modliszewski JL, Gao J, Jung SH, Kirsch DG, Badea CT. Neoadjuvant Radiation Therapy and Surgery Improves Metastasis-Free Survival over Surgery Alone in a Primary Mouse Model of Soft Tissue Sarcoma. Mol Cancer Ther 2023; 22:112-122. [PMID: 36162051 PMCID: PMC9812921 DOI: 10.1158/1535-7163.mct-21-0991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 06/28/2022] [Accepted: 09/20/2022] [Indexed: 02/03/2023]
Abstract
This study aims to investigate whether adding neoadjuvant radiotherapy (RT), anti-programmed cell death protein-1 (PD-1) antibody (anti-PD-1), or RT + anti-PD-1 to surgical resection improves disease-free survival for mice with soft tissue sarcomas (STS). We generated a high mutational load primary mouse model of STS by intramuscular injection of adenovirus expressing Cas9 and guide RNA targeting Trp53 and intramuscular injection of 3-methylcholanthrene (MCA) into the gastrocnemius muscle of wild-type mice (p53/MCA model). We randomized tumor-bearing mice to receive isotype control or anti-PD-1 antibody with or without radiotherapy (20 Gy), followed by hind limb amputation. We used micro-CT to detect lung metastases with high spatial resolution, which was confirmed by histology. We investigated whether sarcoma metastasis was regulated by immunosurveillance by lymphocytes or tumor cell-intrinsic mechanisms. Compared with surgery with isotype control antibody, the combination of anti-PD-1, radiotherapy, and surgery improved local recurrence-free survival (P = 0.035) and disease-free survival (P = 0.005), but not metastasis-free survival. Mice treated with radiotherapy, but not anti-PD-1, showed significantly improved local recurrence-free survival and metastasis-free survival over surgery alone (P = 0.043 and P = 0.007, respectively). The overall metastasis rate was low (∼12%) in the p53/MCA sarcoma model, which limited the power to detect further improvement in metastasis-free survival with addition of anti-PD-1 therapy. Tail vein injections of sarcoma cells into immunocompetent mice suggested that impaired metastasis was due to inability of sarcoma cells to grow in the lungs rather than a consequence of immunosurveillance. In conclusion, neoadjuvant radiotherapy improves metastasis-free survival after surgery in a primary model of STS.
Collapse
Affiliation(s)
- Rutulkumar Patel
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | - Yvonne M. Mowery
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
- Department of Head and Neck Surgery & Communication Sciences, Duke University Medical Center, Durham, NC 27710
| | - Yi Qi
- Department of Radiology, Duke University Medical Center, Durham, NC 27710
| | - Alex M. Bassil
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | - Matt Holbrook
- Department of Radiology, Duke University Medical Center, Durham, NC 27710
| | - Eric S. Xu
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | - Cierra S. Hong
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | - Jonathon E. Himes
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | - Nerissa T. Williams
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | - Jeffrey Everitt
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710
| | - Yan Ma
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | - Lixia Luo
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
| | | | | | - Junheng Gao
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC
| | - Sin-Ho Jung
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC
| | - David G. Kirsch
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708 USA
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710
| | - Cristian T. Badea
- Department of Radiology, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
8
|
Singh G, Sharma SK, Singh SK. miR-34a negatively regulates cell cycle factor Cdt2/DTL in HPV infected cervical cancer cells. BMC Cancer 2022; 22:777. [PMID: 35840896 PMCID: PMC9288023 DOI: 10.1186/s12885-022-09879-5] [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: 04/05/2022] [Accepted: 07/04/2022] [Indexed: 11/10/2022] Open
Abstract
MicroRNAs have emerged as an important regulator of cell cycle and various other cellular processes. Aberration in microRNAs has been linked with development of several cancers and other diseases but still very little is known about the mechanism by which they regulate these cellular events. High risk human papilloma virus (HR HPV) is the causative agent of 99% of cervical cancer cases which attenuates multiple tumor suppressors and checkpoint factors of the host cell. The viral proteins also stabilize many oncogenic factors, including an essential cell cycle regulator Cdt2/DTL which in turn promotes cell transformation and proliferation. In this study, we report that a micro-RNA, miR-34a by suppressing HPV E6 protein, destabilizes Cdt2/DTL protein level in HPV infected cervical cancer cell lines. Destabilization of Cdt2 stabilizes pro-apoptotic and onco-suppressor proteins like p21 and Set8 and suppresses cell proliferation, invasion and migration capabilities of the HPV positive cervical cancer cells. Overexpression of either HPV E6 or Cdt2 genes along with miR-34a restored back the suppressed proliferation rate. This study is the first-ever report to show that miR-34a regulates cell cycle factor Cdt2 by suppressing viral E6 protein level, thus opening up the possibility of exploring miR-34a as a specific therapy for cervical cancer treatment.
Collapse
Affiliation(s)
- Garima Singh
- Cell Cycle and Cancer Laboratory, School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, UP-221005, India
| | - Sonika Kumari Sharma
- Cell Cycle and Cancer Laboratory, School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, UP-221005, India
| | - Samarendra Kumar Singh
- Cell Cycle and Cancer Laboratory, School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, UP-221005, India.
| |
Collapse
|
9
|
Wang Z, Heid B, Lu R, Sachdeva M, Edwards MR, Ren J, Cecere TE, Khan D, Jeboda T, Kirsch DG, Reilly CM, Dai R, Ahmed SA. Deletion of microRNA-183-96-182 Cluster in Lymphocytes Suppresses Anti-DsDNA Autoantibody Production and IgG Deposition in the Kidneys in C57BL/6-Fas lpr/lpr Mice. Front Genet 2022; 13:840060. [PMID: 35873462 PMCID: PMC9301314 DOI: 10.3389/fgene.2022.840060] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Dysregulated miRNAs have been implicated in the pathogenesis of systemic lupus erythematosus (SLE). Our previous study reported a substantial increase in three miRNAs located at the miR-183-96-182 cluster (miR-183C) in several autoimmune lupus-prone mice, including MRL/lpr and C57BL/6-lpr (B6/lpr). This study reports that in vitro inhibition of miR-182 alone or miR-183C by specific antagomirs in activated splenocytes from autoimmune-prone MRL/lpr and control MRL mice significantly reduced lupus-related inflammatory cytokines, interferon-gamma (IFNγ), and IL-6 production. To further characterize the role of miR-182 and miR-183C cluster in vivo in lupus-like disease and lymphocyte phenotypes, we used hCD2-iCre to generate B6/lpr mice with conditional deletion of miR-182 or miR-183C in CD2+ lymphocytes (miR-182-/-B6/lpr and miR-183C-/-B6/lpr). The miR-182-/-B6/lpr and miR-183C-/-B6/lpr mice had significantly reduced deposition of IgG immunocomplexes in the kidney when compared to their respective littermate controls, although there appeared to be no remarkable changes in renal pathology. Importantly, we observed a significant reduction of serum anti-dsDNA autoantibodies in miR-183C-/-B6/lpr mice after reaching 24 weeks-of age compared to age-matched miR-183Cfl/flB6/lpr controls. In vitro activated splenocytes from miR-182-/-B6/lpr mice and miR-183C-/-B6/lpr mice showed reduced ability to produce lupus-associated IFNγ. Forkhead box O1(Foxo1), a previously validated miR-183C miRNAs target, was increased in the splenic CD4+ cells of miR-182-/-B6/lpr and miR-183C-/-B6/lpr mice. Furthermore, in vitro inhibition of Foxo1 with siRNA in splenocytes from miR-182-/-B6/lpr and miR-183C-/-B6/lpr mice significantly increased IFNγ expression following anti-CD3/CD28 stimulation, suggesting that miR-182 and miR-183C miRNAs regulate the inflammatory IFNγ in splenocytes via targeting Foxo1. The deletion of either miR-182 alone or the whole miR-183C cluster, however, had no marked effect on the composition of T and B cell subsets in the spleens of B6/lpr mice. There were similar percentages of CD4+, CD8+, CD19+, as well as Tregs, follicular helper T (TFH), germinal center B (GCB), and plasma cells in the miR-183C-/-B6/lpr and miR-182-/-B6/lpr mice and their respective littermate controls, miR-183Cfl/flB6/lpr and miR-182fl/flB6/lpr mice. Together, our data demonstrate a role of miR-183C in the regulation of anti-dsDNA autoantibody production in vivo in B6/lpr mice and the induction of IFNγ in in vitro activated splenocytes from B6/lpr mice.
Collapse
Affiliation(s)
- Zhuang Wang
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - Bettina Heid
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - Ran Lu
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - Mohit Sachdeva
- Preclinical Lead Immunology, Spark Theraprutics, Philadelphia, PA, United States
| | - Michael R. Edwards
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - JingJing Ren
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - Thomas E. Cecere
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - Deena Khan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - Taschua Jeboda
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - David G. Kirsch
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, United States
| | - Christopher M. Reilly
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
- Edward Via College of Osteopathic Medicine, Blacksburg, VA, United States
| | - Rujuan Dai
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - S. Ansar Ahmed
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
10
|
Szczepanek J, Skorupa M, Tretyn A. MicroRNA as a Potential Therapeutic Molecule in Cancer. Cells 2022; 11:1008. [PMID: 35326459 PMCID: PMC8947269 DOI: 10.3390/cells11061008] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/11/2022] [Accepted: 03/16/2022] [Indexed: 12/11/2022] Open
Abstract
Small noncoding RNAs, as post-translational regulators of many target genes, are not only markers of neoplastic disease initiation and progression, but also markers of response to anticancer therapy. Hundreds of miRNAs have been identified as biomarkers of drug resistance, and many have demonstrated the potential to sensitize cancer cells to therapy. Their properties of modulating the response of cells to therapy have made them a promising target for overcoming drug resistance. Several methods have been developed for the delivery of miRNAs to cancer cells, including introducing synthetic miRNA mimics, DNA plasmids containing miRNAs, and small molecules that epigenetically alter endogenous miRNA expression. The results of studies in animal models and preclinical studies for solid cancers and hematological malignancies have confirmed the effectiveness of treatment protocols using microRNA. Nevertheless, the use of miRNAs in anticancer therapy is not without limitations, including the development of a stable nanoconstruct, delivery method choices, and biodistribution. The aim of this review was to summarize the role of miRNAs in cancer treatment and to present new therapeutic concepts for these molecules. Supporting anticancer therapy with microRNA molecules has been verified in numerous clinical trials, which shows great potential in the treatment of cancer.
Collapse
Affiliation(s)
- Joanna Szczepanek
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Ul. Wilenska 4, 87-100 Torun, Poland;
| | - Monika Skorupa
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Ul. Wilenska 4, 87-100 Torun, Poland;
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Ul. Lwowska 1, 87-100 Torun, Poland;
| | - Andrzej Tretyn
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Ul. Lwowska 1, 87-100 Torun, Poland;
| |
Collapse
|
11
|
Yang W, Yin Y, Bi L, Wang Y, Yao J, Xu L, Jiao L. MiR-182-5p promotes the Metastasis and Epithelial-mesenchymal Transition in Non-small Cell Lung Cancer by Targeting EPAS1. J Cancer 2021; 12:7120-7129. [PMID: 34729113 PMCID: PMC8558643 DOI: 10.7150/jca.60419] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 09/21/2021] [Indexed: 01/17/2023] Open
Abstract
Background: Dysregulation of microRNAs (miRNAs) is associated with the pathogenesis of non-small cell lung cancer (NSCLC). However, the mechanisms through which miR-182-5p regulate NSCLC progression have not been established. This study aimed at evaluating the expression levels of miR-182-5p in human NSCLC and its function in lung cancer cells. Endothelial PAS Domain-containing protein 1 (EPAS1; also referred to as hypoxia-inducing factor 2A, HIF-2α) is a transcription factor that is responsible for induction of genes related to cell survival under hypoxia conditions. Hypoxia, an inherent feature of solid tumors, is associated with aggressive phenotypes, as well as resistance to radiotherapy and chemotherapy, which predict metastasis and poor prognosis. Methods: The Cancer Genome Atlas (TCGA) dataset was used to investigate the association between miR-182-5p expression and clinicopathological characteristics as well as prognosis of NSCLC patients. Target genes of miR-182-5p were identified using the PITA, miRmap, microT, miRanda, PicTar, and TargetScan prediction tools. Transwell assays were performed to determine the potential functions of miR-182-5p in lung cancer cells. Luciferase reporter assays were performed to analyze regulation of the putative target of miR-182-5p while western blot assays were used to validate the luciferase results. Results: miR-182-5p was found to be upregulated in NSCLC tissues and acted as an independent prognostic factor for tumor recurrence in NSCLC patients. Functionally, overexpression of miR-182-5p promoted lung cancer cell migration and invasion. Genome-wide gene expression analysis and luciferase report assays revealed that EPAS1 is a direct target of miR-182-5p. EPAS1 was negatively correlated with miR-182-5p expression in NSCLC tissues. Univariate and multivariate survival analyses identified EPAS1 as an independent prognostic factor for overall survival (OS) in NSCLC. Conclusions: These findings imply that miR-182-5p promotes NSCLC progression by targeting EPAS1 and is, therefore, a potential indicator of tumor recurrence in NSCLC patients.
Collapse
Affiliation(s)
- Wenxiao Yang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yinan Yin
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling Bi
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yichao Wang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jialin Yao
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling Xu
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Cancer Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lijing Jiao
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Clinical Immunology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Damerell V, Pepper MS, Prince S. Molecular mechanisms underpinning sarcomas and implications for current and future therapy. Signal Transduct Target Ther 2021; 6:246. [PMID: 34188019 PMCID: PMC8241855 DOI: 10.1038/s41392-021-00647-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 04/18/2021] [Accepted: 05/18/2021] [Indexed: 02/06/2023] Open
Abstract
Sarcomas are complex mesenchymal neoplasms with a poor prognosis. Their clinical management is highly challenging due to their heterogeneity and insensitivity to current treatments. Although there have been advances in understanding specific genomic alterations and genetic mutations driving sarcomagenesis, the underlying molecular mechanisms, which are likely to be unique for each sarcoma subtype, are not fully understood. This is in part due to a lack of consensus on the cells of origin, but there is now mounting evidence that they originate from mesenchymal stromal/stem cells (MSCs). To identify novel treatment strategies for sarcomas, research in recent years has adopted a mechanism-based search for molecular markers for targeted therapy which has included recapitulating sarcomagenesis using in vitro and in vivo MSC models. This review provides a comprehensive up to date overview of the molecular mechanisms that underpin sarcomagenesis, the contribution of MSCs to modelling sarcomagenesis in vivo, as well as novel topics such as the role of epithelial-to-mesenchymal-transition (EMT)/mesenchymal-to-epithelial-transition (MET) plasticity, exosomes, and microRNAs in sarcomagenesis. It also reviews current therapeutic options including ongoing pre-clinical and clinical studies for targeted sarcoma therapy and discusses new therapeutic avenues such as targeting recently identified molecular pathways and key transcription factors.
Collapse
Affiliation(s)
- Victoria Damerell
- Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, South Africa
| | - Michael S Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology, SAMRC Extramural Unit for Stem Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Sharon Prince
- Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, South Africa.
| |
Collapse
|
13
|
Xiao D, Xie J, Wu S. MicroRNA-182 is a potential biomarker for prognosis of gastric cancer: A protocol for meta-analysis and bioinformatics analysis. Medicine (Baltimore) 2021; 100:e25830. [PMID: 34087825 PMCID: PMC8183740 DOI: 10.1097/md.0000000000025830] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Being the second leading cause of cancer death in the world, gastric cancer is a common malignant tumor in digestive system. Most patients were diagnosed in advanced stage and had poor prognosis. In recent years, related studies have displayed that MicroRNA-182 (miRNA-182) can promote the proliferation, infiltration, metastasis and drug resistance of tumor cells, so it can be used as a new molecular marker for the early diagnosis, prognosis, and treatment of tumors. However, the expression and prognosis of miRNA-182 in gastric cancer are not clear. Therefore, this study conducted a meta-analysis to further clarify the relationship between the expression of miRNA-182 in gastric cancer and prognosis. In addition, a bioinformatics analysis was adopted to further analyze the possible molecular mechanism of miRNA-182, so as to provide a theoretical basis for the diagnosis, treatment, and prognosis of patients suffering from gastric cancer. METHODS The following electronic databases were searched on computer: Wanfang, Chinese Biomedical Literature Database, Chinese National Knowledge Infrastructure, the Chongqing VIP Chinese Science and Technology Periodical Database, PubMed, Embase, and Web of Science databases. The retrieval time is set to build the database until April 2021. Combined hazard ratios (HRs) and 95% confidence intervals (95% CIs) were used to evaluate the effects of miRNA-182 on the prognosis of gastric cancer. Stata 16.0 software was applied for the meta-analysis. The expression of miRNA-182 in gastric cancer was analyzed by Gene Expression Omnibus database and The Cancer Genome Atlas database. The survival curve of miRNA-182 differential expression was analyzed by OncomiR. The target genes of miRNA-182 were predicted by TargetScan, miRBase, miRTarBase, starBase V2.0, and miRWalk. The target genes were obtained by the intersection of Wayne diagram. DAVID database was used for gene ontology (GO) and Kyoto encyclopedia of genes and genomes enrichment analysis. STRING database and Cytoscape were applied to construct Protein-protein interaction network to obtain key genes (hub gene). The expression of hub gene in gastric cancer was analyzed by gene expression profiling interactive analysis. The survival curve between hub gene and prognosis of gastric cancer was drawn by Kaplan-Meier Plotter database. TIMER database was used to analyze the relationship between hub gene expression and immune cell infiltration in gastric cancer. RESULTS The results of this meta-analysis will be submitted to a peer-reviewed journal for publication. CONCLUSION This study provides high-quality evidence support for the expression of miRNA-182 and the prognosis of gastric cancer. Through bioinformatics analysis, we further discussed the mechanism of miRNA-182 in gastric cancer and the understanding of related pathways. OSF REGISTRATION NUMBER DOI 10.17605/OSF.IO/EHJ6X.
Collapse
|
14
|
Galuppini F, Censi S, Moro M, Carraro S, Sbaraglia M, Iacobone M, Fassan M, Mian C, Pennelli G. MicroRNAs in Medullary Thyroid Carcinoma: A State of the Art Review of the Regulatory Mechanisms and Future Perspectives. Cells 2021; 10:955. [PMID: 33924120 PMCID: PMC8074316 DOI: 10.3390/cells10040955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/17/2022] Open
Abstract
Medullary thyroid carcinoma (MTC) is a rare malignant neoplasia with a variable clinical course, with complete remission often difficult to achieve. Genetic alterations lead to fundamental changes not only in hereditary MTC but also in the sporadic form, with close correlations between mutational status and prognosis. In recent years, microRNAs (miRNAs) have become highly relevant as crucial players in MTC etiology. Current research has focused on their roles in disease carcinogenesis and development, but recent studies have expounded their potential as biomarkers and response predictors to novel biological drugs for advanced MTC. One such element which requires greater investigation is their mechanism of action and the molecular pathways involved in the regulation of gene expression. A more thorough understanding of these mechanisms will help realize the promising potential of miRNAs for MTC therapy and management.
Collapse
Affiliation(s)
- Francesca Galuppini
- Pathology Unit, Department of Medicine, University of Padua, Via Gabelli 61, 35121 Padua, Italy; (F.G.); (M.M.); (S.C.); (M.S.); (M.F.)
| | - Simona Censi
- Endocrinology Unit, Department of Medicine, University of Padua, Via Ospedale Civile 105, 35121 Padua, Italy; (S.C.); (C.M.)
| | - Margherita Moro
- Pathology Unit, Department of Medicine, University of Padua, Via Gabelli 61, 35121 Padua, Italy; (F.G.); (M.M.); (S.C.); (M.S.); (M.F.)
| | - Stefano Carraro
- Pathology Unit, Department of Medicine, University of Padua, Via Gabelli 61, 35121 Padua, Italy; (F.G.); (M.M.); (S.C.); (M.S.); (M.F.)
| | - Marta Sbaraglia
- Pathology Unit, Department of Medicine, University of Padua, Via Gabelli 61, 35121 Padua, Italy; (F.G.); (M.M.); (S.C.); (M.S.); (M.F.)
| | - Maurizio Iacobone
- Endocrine Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Giustiniani 2, 35128 Padua, Italy;
| | - Matteo Fassan
- Pathology Unit, Department of Medicine, University of Padua, Via Gabelli 61, 35121 Padua, Italy; (F.G.); (M.M.); (S.C.); (M.S.); (M.F.)
- Istituto Oncologico del Veneto, IOV-IRCCS, 35128 Padova, Italy
| | - Caterina Mian
- Endocrinology Unit, Department of Medicine, University of Padua, Via Ospedale Civile 105, 35121 Padua, Italy; (S.C.); (C.M.)
| | - Gianmaria Pennelli
- Pathology Unit, Department of Medicine, University of Padua, Via Gabelli 61, 35121 Padua, Italy; (F.G.); (M.M.); (S.C.); (M.S.); (M.F.)
| |
Collapse
|
15
|
Grzywa TM, Klicka K, Włodarski PK. Regulators at Every Step-How microRNAs Drive Tumor Cell Invasiveness and Metastasis. Cancers (Basel) 2020; 12:E3709. [PMID: 33321819 PMCID: PMC7763175 DOI: 10.3390/cancers12123709] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor cell invasiveness and metastasis are the main causes of mortality in cancer. Tumor progression is composed of many steps, including primary tumor growth, local invasion, intravasation, survival in the circulation, pre-metastatic niche formation, and metastasis. All these steps are strictly controlled by microRNAs (miRNAs), small non-coding RNA that regulate gene expression at the post-transcriptional level. miRNAs can act as oncomiRs that promote tumor cell invasion and metastasis or as tumor suppressor miRNAs that inhibit tumor progression. These miRNAs regulate the actin cytoskeleton, the expression of extracellular matrix (ECM) receptors including integrins and ECM-remodeling enzymes comprising matrix metalloproteinases (MMPs), and regulate epithelial-mesenchymal transition (EMT), hence modulating cell migration and invasiveness. Moreover, miRNAs regulate angiogenesis, the formation of a pre-metastatic niche, and metastasis. Thus, miRNAs are biomarkers of metastases as well as promising targets of therapy. In this review, we comprehensively describe the role of various miRNAs in tumor cell migration, invasion, and metastasis.
Collapse
Affiliation(s)
- Tomasz M. Grzywa
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Klaudia Klicka
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Paweł K. Włodarski
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
| |
Collapse
|
16
|
Huang J, Sachdeva M, Xu E, Robinson TJ, Luo L, Ma Y, Williams NT, Lopez O, Cervia LD, Yuan F, Qin X, Zhang D, Owzar K, Gokgoz N, Seto A, Okada T, Singer S, Andrulis IL, Wunder JS, Lazar AJ, Rubin BP, Pipho K, Mello SS, Giudice J, Kirsch DG. The Long Noncoding RNA NEAT1 Promotes Sarcoma Metastasis by Regulating RNA Splicing Pathways. Mol Cancer Res 2020; 18:1534-1544. [PMID: 32561656 PMCID: PMC7541426 DOI: 10.1158/1541-7786.mcr-19-1170] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/09/2020] [Accepted: 06/15/2020] [Indexed: 11/16/2022]
Abstract
Soft-tissue sarcomas (STS) are rare malignancies showing lineage differentiation toward diverse mesenchymal tissues. Half of all high-grade STSs develop lung metastasis with a median survival of 15 months. Here, we used a genetically engineered mouse model that mimics undifferentiated pleomorphic sarcoma (UPS) to study the molecular mechanisms driving metastasis. High-grade sarcomas were generated with Cre recombinase technology using mice with conditional mutations in Kras and Trp53 (KP) genes. After amputation of the limb bearing the primary tumor, mice were followed for the development of lung metastasis. Using RNA-sequencing of matched primary KP tumors and lung metastases, we found that the long noncoding RNA (lncRNA) Nuclear Enriched Abundant Transcript 1 (Neat1) is significantly upregulated in lung metastases. Furthermore, NEAT1 RNA ISH of human UPS showed that NEAT1 is upregulated within a subset of lung metastases compared with paired primary UPS. Remarkably, CRISPR/Cas9-mediated knockout of Neat1 suppressed the ability of KP tumor cells to colonize the lungs. To gain insight into the underlying mechanisms by which the lncRNA Neat1 promotes sarcoma metastasis, we pulled down Neat1 RNA and used mass spectrometry to identify interacting proteins. Interestingly, most Neat1 interacting proteins are involved in RNA splicing regulation. In particular, KH-Type Splicing Regulatory Protein (KHSRP) interacts with Neat1 and is associated with poor prognosis of human STS. Moreover, depletion of KHSRP suppressed the ability of KP tumor cells to colonize the lungs. Collectively, these results suggest that Neat1 and its interacting proteins, which regulate RNA splicing, are involved in mediating sarcoma metastasis. IMPLICATIONS: Understanding that lncRNA NEAT1 promotes sarcoma metastasis, at least in part, through interacting with the RNA splicing regulator KHSRP may translate into new therapeutic approaches for sarcoma.
Collapse
Affiliation(s)
- Jianguo Huang
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Mohit Sachdeva
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Eric Xu
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Timothy J Robinson
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Nerissa T Williams
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Omar Lopez
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| | - Lisa D Cervia
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Xiaodi Qin
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| | - Dadong Zhang
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| | - Kouros Owzar
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
- Department of Biostatistics & Bioinformatics, Duke University, Durham, North Carolina
| | - Nalan Gokgoz
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Andrew Seto
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Tomoyo Okada
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Samuel Singer
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Irene L Andrulis
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Jay S Wunder
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- University of Toronto Musculoskeletal Oncology Unit, and Department of Surgery, University of Toronto, Toronto, Canada
| | - Alexander J Lazar
- Departments of Pathology, Genomic Medicine, and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Brian P Rubin
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Krista Pipho
- University of Rochester Medical Center, Rochester, New York
| | | | - Jimena Giudice
- Department of Cell Biology and Physiology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- McAllister Heart Institute, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - David G Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina.
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
17
|
Tang YJ, Huang J, Tsushima H, Ban GI, Zhang H, Oristian KM, Puviindran V, Williams N, Ding X, Ou J, Jung SH, Lee CL, Jiao Y, Chen BJ, Kirsch DG, Alman BA. Tracing Tumor Evolution in Sarcoma Reveals Clonal Origin of Advanced Metastasis. Cell Rep 2020; 28:2837-2850.e5. [PMID: 31509746 PMCID: PMC6750751 DOI: 10.1016/j.celrep.2019.08.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/17/2019] [Accepted: 08/05/2019] [Indexed: 11/03/2022] Open
Abstract
Cellular heterogeneity is frequently observed in cancer, but the biological significance of heterogeneous tumor clones is not well defined. Using multicolor reporters and CRISPR-Cas9 barcoding, we trace clonal dynamics in a mouse model of sarcoma. We show that primary tumor growth is associated with a reduction in clonal heterogeneity. Local recurrence of tumors following surgery or radiation therapy is driven by multiple clones. In contrast, advanced metastasis to the lungs is driven by clonal selection of a single metastatic clone (MC). Using RNA sequencing (RNA-seq) and in vivo assays, we identify candidate suppressors of metastasis, namely, Rasd1, Reck, and Aldh1a2. These genes are downregulated in MCs of the primary tumors prior to the formation of metastases. Overexpression of these suppressors of metastasis impair the ability of sarcoma cells to colonize the lungs. Overall, this study reveals clonal dynamics during each step of tumor progression, from initiation to growth, recurrence, and distant metastasis.
Collapse
Affiliation(s)
- Yuning J Tang
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jianguo Huang
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Hidetoshi Tsushima
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Ga I Ban
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Hongyuan Zhang
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA; Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Kristianne M Oristian
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Vijitha Puviindran
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Nerissa Williams
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Xiruo Ding
- Department of Biostatics and Bioinformatics, Duke University, Durham, NC, USA
| | - Jianhong Ou
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA; Regeneration Next Initiative, Duke University, Durham, NC, USA
| | - Sin-Ho Jung
- Department of Biostatics and Bioinformatics, Duke University, Durham, NC, USA
| | - Chang-Lung Lee
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Yiqun Jiao
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Benny J Chen
- Duke Cancer Institute, Duke University, Durham, NC, USA; Regeneration Next Initiative, Duke University, Durham, NC, USA
| | - David G Kirsch
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA; Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA; Duke Cancer Institute, Duke University, Durham, NC, USA; Regeneration Next Initiative, Duke University, Durham, NC, USA.
| | - Benjamin A Alman
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA; Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA; Duke Cancer Institute, Duke University, Durham, NC, USA; Regeneration Next Initiative, Duke University, Durham, NC, USA; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
18
|
MicroRNA-Dependent Targeting of RSU1 and the IPP Adhesion Complex Regulates the PTEN/PI3K/AKT Signaling Pathway in Breast Cancer Cell Lines. Int J Mol Sci 2020; 21:ijms21155458. [PMID: 32751711 PMCID: PMC7432699 DOI: 10.3390/ijms21155458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/21/2022] Open
Abstract
(1) Background: The microRNA (miR)-directed control of gene expression is correlated with numerous physiological processes as well as the pathological features of tumors. The focus of this study is on the role of miRs in the regulation of RSU1 and proteins in the IPP (integrin linked kinase, PINCH and parvin) complex. Because the IPP adaptor proteins link β integrins to actin cytoskeleton, and the RSU1 signaling protein connects the complex to the activation of cJun, ATF2 and the transcription of PTEN, their reduction by miRs has the potential to alter both adhesion and survival signaling. (2) Methods: Multiple database analyses were used to identify miRs that target RSU1 and PINCH1. miR transfection validated the effects of miRs on RSU1, PINCH1 and downstream targets in breast cancer cell lines. (3) Results: The miRs targeting RSU1 mRNA include miR-182-5p, -409-3p, -130a-3p, -221-3p, -744-5p and -106b-5p. Data show that miR-182-5p and -409-3p reduce RSU1, PINCH1 and inhibit the ATF2 activation of PTEN expression. miR-221-3p and miR-130a-3p target RSU1 and PINCH1 and, conversely, RSU1 depletion increases miR-221-3p and miR-130a-3p. (4) Conclusions: miRs targeting RSU1 and PINCH1 in mammary epithelial or luminal breast cancer cell lines reduced RSU1 signaling to p38 MAP kinase and ATF2, inhibiting the expression of PTEN. miR-221-3p, known to target PTEN and cell cycle regulators, also targets RSU1 and PINCH1 in luminal breast cancer cell lines.
Collapse
|
19
|
Lu JT, Tan CC, Wu XR, He R, Zhang X, Wang QS, Li XQ, Zhang R, Feng YM. FOXF2 deficiency accelerates the visceral metastasis of basal-like breast cancer by unrestrictedly increasing TGF-β and miR-182-5p. Cell Death Differ 2020; 27:2973-2987. [PMID: 32424142 DOI: 10.1038/s41418-020-0555-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/15/2022] Open
Abstract
The mesenchymal transcription factor forkhead box F2 (FOXF2) is a critical regulator of embryogenesis and tissue homeostasis. Our previous studies demonstrated that FOXF2 is ectopically expressed in basal-like breast cancer (BLBC) cells and that FOXF2 deficiency promotes the epithelial-mesenchymal transition and aggressiveness of BLBC cells. In this study, we found that FOXF2 controls transforming growth factor-beta (TGF-β)/SMAD signaling pathway activation through transrepression of TGF-β-coding genes in BLBC cells. FOXF2-deficient BLBC cells adopt a myofibroblast-/cancer-associated fibroblast (CAF)-like phenotype, and tend to metastasize to visceral organs by increasing autocrine TGF-β signaling and conferring aggressiveness to neighboring cells by increasing paracrine TGF-β signaling. In turn, TGF-β silences FOXF2 expression through upregulating miR-182-5p, a posttranscriptional regulator of FOXF2 and inducer of metastasis. In addition to mediating a reciprocal repression loop between FOXF2 and TGF-β through direct transrepression by SMAD3, miR-182-5p forms a reciprocal repression loop with FOXF2 that directly transrepresses MIR182 expression. Therefore, FOXF2 deficiency accelerates the visceral metastasis of BLBC through unrestricted increases in autocrine and paracrine TGF-β signaling, and miR-182-5p expression. Our findings provide novel mechanisms underlying the roles of TGF-β, miR-182-5p, and FOXF2 in accelerating BLBC dissemination and metastasis, and may facilitate the development of therapeutic strategies for aggressive BLBC.
Collapse
Affiliation(s)
- Jun-Tao Lu
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Cong-Cong Tan
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Xiao-Ran Wu
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Rui He
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Xiao Zhang
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Qing-Shan Wang
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Treatment of the Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Xiao-Qing Li
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Treatment of the Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Rui Zhang
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Treatment of the Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Yu-Mei Feng
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China. .,Key Laboratory of Breast Cancer Prevention and Treatment of the Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China.
| |
Collapse
|
20
|
Inoue K, Hu X, Zhao B. Regulatory network mediated by RBP-J/NFATc1-miR182 controls inflammatory bone resorption. FASEB J 2020; 34:2392-2407. [PMID: 31908034 PMCID: PMC7018544 DOI: 10.1096/fj.201902227r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/21/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022]
Abstract
Bone resorption is a severe consequence of inflammatory diseases associated with osteolysis, such as rheumatoid arthritis (RA), often leading to disability in patients. In physiological conditions, the differentiation of bone-resorbing osteoclasts is delicately regulated by the balance between osteoclastogenic and anti-osteoclastogenic mechanisms. Inflammation has complex impact on osteoclastogenesis and bone destruction, and the underlying mechanisms of which, especially feedback inhibition, are underexplored. Here, we identify a novel regulatory network mediated by RBP-J/NFATc1-miR182 in TNF-induced osteoclastogenesis and inflammatory bone resorption. This network includes negative regulator RBP-J and positive regulators, NFATc1 and miR182, of osteoclast differentiation. In this network, miR182 is a direct target of both RBP-J and NFATc1. RBP-J represses, while NFATc1 activates miR182 expression through binding to specific open chromatin regions in the miR182 promoter. Inhibition of miR182 by RBP-J servers as a critical mechanism that limits TNF-induced osteoclast differentiation and inflammatory bone resorption. Inflammation, such as that which occurs in RA, shifts the expression levels of the components in this network mediated by RBP-J/NFATc1-miR182-FoxO3/PKR (previously identified miR182 targets) towards more osteoclastogenic, rather than healthy conditions. Treatment with TNF inhibitors in RA patients reverses the expression changes of the network components and osteoclastogenic potential. Thus, this network controls the balance between activating and repressive signals that determine the extent of osteoclastogenesis. These findings collectively highlight the biological significance and translational implication of this newly identified intrinsic regulatory network in inflammatory osteoclastogenesis and osteolysis.
Collapse
Affiliation(s)
- Kazuki Inoue
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Xiaoyu Hu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
- Graduate Program in Cell and Development Biology, Weill Cornell Graduate School of Medical Sciences of Cornell University, New York, New York, USA
| |
Collapse
|
21
|
Orso F, Quirico L, Dettori D, Coppo R, Virga F, Ferreira LC, Paoletti C, Baruffaldi D, Penna E, Taverna D. Role of miRNAs in tumor and endothelial cell interactions during tumor progression. Semin Cancer Biol 2020; 60:214-224. [DOI: 10.1016/j.semcancer.2019.07.024] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 07/29/2019] [Indexed: 12/18/2022]
|
22
|
Yao XG, Tan Q, Liu PP, Feng LJ. Tissue microRNA-182 expression level and its potential prognostic value for papillary thyroid carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:3128-3133. [PMID: 31934155 PMCID: PMC6949707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/23/2015] [Indexed: 06/10/2023]
Abstract
BACKGROUND In previous study, qRT-PCR analysis revealed significantly higher miR-182 levels in papillary thyroid carcinoma (PTC) than matched normal tissues. However, the clinical significance and prognostic value of miR-182 have not been investigated in PTC until now. METHODS 151 pairs of PTC and adjacent normal thyroid tissues were obtained from Affiliated Hospital of Weifang Medical University from February 2008 to January 2015. The Chi square test was used to analyze the relationship between miR-182 expression and the clinicopathological characteristics. We used the Kaplan-Meier method and the log-rank test in univariate survival analysis, and we used the Cox proportional hazards regression model in our multivariate analysis. RESULTS The relative expression of miR-182 in PTC samples was significantly higher than that of matched normal tissues (P<0.001). The high expression level of tissue miR-182 was statistically correlated with extrathyroidal invasion (P=0.009), cervical lymphnode metastasis (P=0.015), and TNM staging (P=0.001). The Kaplan-Meier method revealed that higher miR-182 expression level was correlated with significantly reduced overall survival. Furthermore, multivariate survival analysis revealed that miR-182 expression level (HR=2.882, 95% CI: 1.289-10.928, P=0.013) was significantly correlated with the poor prognosis of PTC patients. CONCLUSIONS Overexpression of miR-182 is associated with aggressive clinicopathologic characteristics of PTC, and miR-182 might be a novel prognostic molecular marker of PTC.
Collapse
Affiliation(s)
- Xing-Guo Yao
- Department of Hepatobiliary Surgery, Affiliated Hospital of Weifang Medical UniversityWeifang 261031, Shandong Province, China
| | - Qiang Tan
- Department of Respiratory Medicine, Weifang People’s HospitalWeifang 261041, Shandong Province, China
| | - Ping-Ping Liu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Weifang Medical UniversityWeifang 261031, Shandong Province, China
| | - Ling-Jun Feng
- Department of Thyroid & Breast Surgery, Affiliated Hospital of Weifang Medical UniversityWeifang 261031, Shandong Province, China
| |
Collapse
|
23
|
Barnaś E, Raś R, Skręt-Magierło J, Wesecki M, Filipowska J, Książek M, Skręt A, Widenka K. Natural history of leiomyomas beyond the uterus. Medicine (Baltimore) 2019; 98:e15877. [PMID: 31232922 PMCID: PMC6636938 DOI: 10.1097/md.0000000000015877] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
RATIONALE Most leiomyomas are located in the uterus. Leiomyomas are rarely found outside the uterus and classified as leiomyoma beyond the uterus (LBU). This group consists of disseminated peritoneal leiomyomatosis, benign metastasizing leiomyoma, intravenous leiomyomatosis, parasite leiomyoma located in the broad ligament and retroperitoneal space. The descriptions of the patients who suffer from these types of leiomyomas are presented mainly in case reports. PATIENT CONCERNS A 34-year-old multiparous woman was operated on multiple recurrent uterine leiomyoma in parametrium. At one time, 32 leiomyomas were removed. Thirteen months following it, in next laparotomy, 132 leiomyomas were excised. Histologically, both were intravenous leiomyomas (IVLs). DIAGNOSIS AND INTERVENTIONS In follow-up, computed tomography (CT) and magnetic resonance imaging scans were performed to look for next recurrent leiomyoma. Accidentally, the mass was found in inferior vena cava which was diagnosed as intravenous vena cava leiomyoma. The mass was removed and the final diagnosis of intravenous myoma was confirmed in histopathology. OUTCOMES CT scan performed 3 months after the surgery for leiomyoma in vena cava revealed no pathology. Next 10 months' follow-up was uneventful. LESSONS The recurrent multiple uterine leiomyoma precede LBU. The uterine leiomyoma spreads intravenously route to parametria as parasite leiomyoma, then to vena cava. It has to be taken into account in follow-up.
Collapse
Affiliation(s)
| | - Renata Raś
- Department of Physics, Rzeszow University of Technology
| | | | - Mariusz Wesecki
- Oncology Surgery Department, Specialist Hospital in Brzozów, Podkarpacki Oncological Center, Brzozów
| | - Justyna Filipowska
- Chair of Electroradiology, Institute of Nursing and Health Sciences, Faculty of Medicine, University of Rzeszow
| | - Mariusz Książek
- Clinical Department of Pathology, Frederick Chopin Clinical Provincial Hospital No 1
| | - Andrzej Skręt
- Obstetrics and Gynecology Clinic, Medical Faculty, University of Rzeszow, Rzeszow
| | - Kazimierz Widenka
- Clinical Department of Cardiac Surgery, Medical Faculty, University of Rzeszów, Rzeszów, Poland
| |
Collapse
|
24
|
Coronel-Hernández J, López-Urrutia E, Contreras-Romero C, Delgado-Waldo I, Figueroa-González G, Campos-Parra AD, Salgado-García R, Martínez-Gutierrez A, Rodríguez-Morales M, Jacobo-Herrera N, Terrazas LI, Silva-Carmona A, López-Camarillo C, Pérez-Plasencia C. Cell migration and proliferation are regulated by miR-26a in colorectal cancer via the PTEN-AKT axis. Cancer Cell Int 2019; 19:80. [PMID: 30983885 PMCID: PMC6444875 DOI: 10.1186/s12935-019-0802-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/23/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Invasion and metastasis are determinant events in the prognosis of Colorectal cancer (CRC), a common neoplasm worldwide. An important factor for metastasis is the acquired capacity of the cell to proliferate and invade adjacent tissues. In this paper, we explored the role of micro-RNA-26a in the regulation of proliferation and migration in CRC-derived cells through the negative regulation of PTEN, a key negative regulator of the AKT pathway. METHODS Expression levels of PTEN and mir-26a were surveyed in normal and CRC-derived cell lines; paraffin embedded human tissues, TCGA CRC expression data and a Balb/c mice orthotopic induced CRC model. CRC was induced by an initial intraperitoneal dose of the colonic carcinogen Azoxymethane followed by inflammatory promoter Dextran Sulfate Sodium Salt. Luciferase assays provide information about miR-26a-PTEN 3'UTR interaction. Proliferation and migration by real time cell analysis and wound-healing functional analyses were performed to assess the participation of mir-26a on important hallmarks of CRC and its regulation on the PTEN gene. RESULTS We observed a negative correlation between PTEN and mir-26a expression in cell lines, human tissues, TCGA data, and tissues derived from the CRC mouse model. Moreover, we showed that negative regulation of PTEN exerted by miR-26a affected AKT phosphorylation levels directly. Functional assays showed that mir-26a directly down-regulates PTEN, and that mir-26a over-expressing cells had higher proliferation and migration rates. CONCLUSIONS All this data proposes an important role of mir-26a as an oncomir in the progression and invasion of CRC. Our data suggested that mir-26a could be used as a biomarker of tumor development in CRC patients, however more studies must be conducted to establish its clinical role.
Collapse
Affiliation(s)
- Jossimar Coronel-Hernández
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, FES-IZTACALA, UNAM, Tlalnepantla, Mexico
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Eduardo López-Urrutia
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, FES-IZTACALA, UNAM, Tlalnepantla, Mexico
| | - Carlos Contreras-Romero
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, FES-IZTACALA, UNAM, Tlalnepantla, Mexico
| | - Izamary Delgado-Waldo
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, FES-IZTACALA, UNAM, Tlalnepantla, Mexico
| | - Gabriela Figueroa-González
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Av. San Fernando No 22, Col. Sección XVI, Tlalpan, Zip code 14080 Mexico City, DF Mexico
| | - Alma D. Campos-Parra
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Av. San Fernando No 22, Col. Sección XVI, Tlalpan, Zip code 14080 Mexico City, DF Mexico
| | - Rebeca Salgado-García
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Av. San Fernando No 22, Col. Sección XVI, Tlalpan, Zip code 14080 Mexico City, DF Mexico
| | - Antonio Martínez-Gutierrez
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Av. San Fernando No 22, Col. Sección XVI, Tlalpan, Zip code 14080 Mexico City, DF Mexico
| | - Miguel Rodríguez-Morales
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Av. San Fernando No 22, Col. Sección XVI, Tlalpan, Zip code 14080 Mexico City, DF Mexico
| | - Nadia Jacobo-Herrera
- Unidad de Bioquímica, Instituto de Ciencias Médicas y Nutrición, Salvador Zubirán, Tlalpan, Mexico City, DF Mexico
| | - Luis Ignacio Terrazas
- Laboratorio de Inmunología de Parásitos, Unidad de Biomedicina, FES-IZTACALA, UNAM, Tlalnepantla, Mexico
| | - Abraham Silva-Carmona
- Laboratorio de Genética, Genómica y Bioinformática, Hospital Infantil de México, Mexico City, Mexico
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico City, Mexico
| | - Carlos Pérez-Plasencia
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, FES-IZTACALA, UNAM, Tlalnepantla, Mexico
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Av. San Fernando No 22, Col. Sección XVI, Tlalpan, Zip code 14080 Mexico City, DF Mexico
| |
Collapse
|
25
|
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs regulating post-transcriptional gene expression. They play important roles in many biological processes under physiological or pathological conditions, including development, metabolism, tumorigenesis, metastasis, and immune response. Over the past 15 years, significant insights have been gained into the roles of miRNAs in cancer. Depending on the cancer type, miRNAs can act as oncogenes, tumor suppressors, or metastasis regulators. In this review, we focus on the role of miRNAs as components of molecular networks regulating metastasis. These miRNAs, termed metastamiRs, promote or inhibit metastasis through various mechanisms, including regulation of migration, invasion, colonization, cancer stem cell properties, epithelial-mesenchymal transition, and microenvironment. Some of these metastamiRs represent attractive therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Jongchan Kim
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Fan Yao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Zhenna Xiao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yutong Sun
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA. .,UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
26
|
Lu L, Liu Q, Wang P, Wu Y, Liu X, Weng C, Fang X, Li B, Cao X, Mao H, Wang L, Guan M, Wang W, Liu G. MicroRNA-148b regulates tumor growth of non-small cell lung cancer through targeting MAPK/JNK pathway. BMC Cancer 2019; 19:209. [PMID: 30849960 PMCID: PMC6408859 DOI: 10.1186/s12885-019-5400-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 02/21/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND MicroRNA-148b (miR-148b) has been detected in various types of tumors, and is generally viewed as a tumor suppressor. Our previous study found the decreased expression of miR-148b in human non small cell lung cancer (NSCLC) specimens and cell lines. However, the underlying mechanisms of miR-148b in regulating tumor progression remain unclear. METHODS Firstly animal experiments were performed to verify whether miR-148b could inhibit the tumor growth. Then, the underlying mechanisms were studied by transfecting recombinant plasmids containing a miR-148b mimic or a negative control (NC) mimic (shRNA control) into NSCLC cell lines PC14/B and A549 cells. Tumor cells transfected with unpackaged lentiviral vectors was used as blank control. Cell proliferation capabilities were measured by using CCK-8 kit and colony formation assay. Cell cycle arrest was compared to clarify the mechanism underlying the tumor cell proliferation. Annexin V-FITC Apoptosis Detection kit was applied to investigate the effect of miR-148b on cell apoptosis. Furthermore, western blot analysis were performed to study the targeting pathway. RESULTS We found that over-expression of miR148b could significantly inhibit tumor growth, while knocking down miR148b could obviously promote tumor growth. Further experiment showed that miR-148b inhibited tumor cell proliferation. Besides, over-expression of miR148b decreased the G2/M phase population of the cell cycle by preventing NSCLC cells from entering the mitotic phase and enhanced tumor cell apoptosis. Further western blot analysis indicated that miR148b could inhibit mitogen-activated protein kinase/Jun N-terminal kinase (MAPK/JNK) signaling by decreasing the expression of phosphorylated (p) JNK. CONCLUSIONS These results demonstrate that miR-148b could inhibit the tumor growth and act as tumor suppressor by inhibiting the proliferation and inducing apoptosis of NSCLC cells by blocking the MAPK/JNK pathway.
Collapse
Affiliation(s)
- Lin Lu
- Department of Medical Oncology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, 510180 Guangdong China
- Department of Medical Oncology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180 Guangdong China
| | - Qiyao Liu
- Department of Medical Oncology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, 510180 Guangdong China
- Department of Medical Oncology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180 Guangdong China
| | - Peipei Wang
- Department of Medical Oncology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, 510180 Guangdong China
- Department of Medical Oncology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180 Guangdong China
| | - Yong Wu
- Department of Medical Oncology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, 510180 Guangdong China
- Department of Medical Oncology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180 Guangdong China
| | - Xia Liu
- Department of Medical Oncology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, 510180 Guangdong China
- Department of Medical Oncology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180 Guangdong China
| | - Chengyin Weng
- Department of Medical Oncology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, 510180 Guangdong China
- Department of Medical Oncology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180 Guangdong China
| | - Xisheng Fang
- Department of Medical Oncology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, 510180 Guangdong China
- Department of Medical Oncology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180 Guangdong China
| | - Baoxiu Li
- Department of Medical Oncology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, 510180 Guangdong China
- Department of Medical Oncology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180 Guangdong China
| | - Xiaofei Cao
- Department of Medical Oncology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, 510180 Guangdong China
- Department of Medical Oncology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180 Guangdong China
| | - Haibo Mao
- Department of Medical Oncology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, 510180 Guangdong China
- Department of Medical Oncology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180 Guangdong China
| | - Lina Wang
- Department of Medical Oncology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, 510180 Guangdong China
- Department of Medical Oncology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180 Guangdong China
| | - Mingmei Guan
- Department of Medical Oncology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, 510180 Guangdong China
- Department of Medical Oncology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180 Guangdong China
| | - Wei Wang
- Department of Experimental Research and State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Guangzhou, 510080 Guangdong China
| | - Guolong Liu
- Department of Medical Oncology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, 510180 Guangdong China
- Department of Medical Oncology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180 Guangdong China
| |
Collapse
|
27
|
Song EJ, Ashcraft KA, Lowery CD, Mowery YM, Luo L, Ma Y, Campos LDS, Cardona DM, Stancato L, Kirsch DG. Investigating a chimeric anti-mouse PDGFRα antibody as a radiosensitizer in primary mouse sarcomas. EBioMedicine 2019; 40:224-230. [PMID: 30711517 PMCID: PMC6413473 DOI: 10.1016/j.ebiom.2019.01.046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/19/2019] [Accepted: 01/22/2019] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Olaratumab (LY3012207/IMC-3G3/Lartruvo™) is a fully human monoclonal antibody specific for platelet-derived growth factor receptor alpha (PDGFRα). Phase Ib/II trial results of olaratumab plus doxorubicin in adult patients with advanced soft tissue sarcoma (STS) supported accelerated FDA approval of this regimen. Radiation therapy (RT) is frequently used for high-risk localized STS. However, olaratumab has not been tested with concurrent RT. Here, we evaluate the chimeric anti-mouse PDGFRα antibody 1E10Fc as a radiosensitizer in a primary mouse model of STS. METHODS Primary STS were initiated in mice. When tumors reached 70 mm3, mice were allocated into treatment groups: 1) isotype, 2) 1E10Fc, 3) isotype + RT, 4) 1E10Fc + RT. 1E10Fc or isotype was given biweekly. RT (25 Gy delivered in 5 daily 5 Gy fractions) was initiated on Day 0 with first drug treatment. Tumors were measured 3× per week. Upon reaching 900 mm3, tumors and lungs were harvested. A two-way ANOVA was performed to compare tumor growth delay. Primary tumors were stained for CD31 and PDGFRα and lungs were assessed for micrometastases. A Chi-square test was performed to compare the development of micrometastases in the lungs after treatment with 1E10Fc or isotype. FINDINGS RT significantly delayed time to tumor quintupling compared to no RT (p < 0·0001) [two-way ANOVA], but no difference in tumor growth was seen between mice receiving isotype or 1E10Fc treatment regardless of concurrent RT. Lower microvessel density was observed in the 1E10Fc + RT group. Fewer mice treated with 1E10Fc had micrometastases, but this difference was not statistically significant (p < 0·09). INTERPRETATION 1E10Fc did not act as a radiosensitizer in this primary STS model. FUNDING This study was funded by a research agreement from Eli Lilly and Company.
Collapse
Affiliation(s)
- Erin J Song
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Kathleen A Ashcraft
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, United States
| | | | - Yvonne M Mowery
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Lixia Luo
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Yan Ma
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Lorraine Da Silva Campos
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Diana M Cardona
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Louis Stancato
- Eli Lilly and Company, Indianapolis, IN 46285, United States
| | - David G Kirsch
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, United States; Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, United States.
| |
Collapse
|
28
|
Mollaei H, Safaralizadeh R, Rostami Z. MicroRNA replacement therapy in cancer. J Cell Physiol 2019; 234:12369-12384. [PMID: 30605237 DOI: 10.1002/jcp.28058] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/17/2018] [Indexed: 12/13/2022]
Abstract
Despite the recent progress in cancer management approaches, the mortality rate of cancer is still growing and there are lots of challenges in the clinics in terms of novel therapeutics. MicroRNAs (miRNA) are regulatory small noncoding RNAs and are already confirmed to have a great role in regulating gene expression level by targeting multiple molecules that affect cell physiology and disease development. Recently, miRNAs have been introduced as promising therapeutic targets for cancer treatment. Regulatory potential of tumor suppressor miRNAs, which enables regulation of entire signaling networks within the cells, makes them an interesting option for developing cancer therapeutics. In this regard, over recent decades, scientists have aimed at developing powerful and safe targeting approaches to restore these suppressive miRNAs in cancerous cells. The present review summarizes the function of miRNAs in tumor development and presents recent findings on how miRNAs have served as therapeutic agents against cancer, with a special focus on tumor suppressor miRNAs (mimics). Moreover, the latest investigations on the therapeutic strategies of miRNA delivery have been presented.
Collapse
Affiliation(s)
- Homa Mollaei
- Department of Biology, Faculty of Sciences, University of Birjand, Birjand, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Zeinab Rostami
- Department of Immunology, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
29
|
Van Mater D, Xu E, Reddy A, Añó L, Sachdeva M, Huang W, Williams N, Ma Y, Love C, Happ L, Dave S, Kirsch DG. Injury promotes sarcoma development in a genetically and temporally restricted manner. JCI Insight 2018; 3:123687. [PMID: 30333301 DOI: 10.1172/jci.insight.123687] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 08/30/2018] [Indexed: 11/17/2022] Open
Abstract
Cancer results from the accumulation of genetic mutations in a susceptible cell of origin. We and others have also shown that injury promotes sarcoma development, but how injury cooperates with genetic mutations at the earliest stages of tumor formation is not known. Here, we utilized dual recombinase technology to dissect the complex interplay of the timing of KrasG12D activation, p53 deletion, and muscle injury in sarcomagenesis using a primary mouse model of soft tissue sarcoma. When mutations in oncogenic Kras and p53 are separated by 3 weeks, few sarcomas develop without injury. However, the transformation potential of these tumor-initiating cells can be unmasked by muscle injury. In the absence of Kras mutations, injury of the muscle with global deletion of p53 results in sarcomas with amplification of chromosomal regions encompassing the Met or Yap1 gene. These findings demonstrate a complex interplay between the timing of genetic mutations and perturbations in the tumor microenvironment, which provides insight into the earliest stages of sarcoma development.
Collapse
Affiliation(s)
| | - Eric Xu
- Department of Radiation Oncology
| | - Anupama Reddy
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, and
| | - Leonor Añó
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | | - Yan Ma
- Department of Radiation Oncology
| | - Cassandra Love
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, and
| | - Lanie Happ
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, and
| | - Sandeep Dave
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, and
| | - David G Kirsch
- Department of Radiation Oncology.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
30
|
Abstract
Targeting microRNAs recently shows significant therapeutic promise; however, such progress is underdeveloped in treatment of skeletal diseases with osteolysis, such as osteoporosis and rheumatoid arthritis (RA). Here, we identified miR-182 as a key osteoclastogenic regulator in bone homeostasis and diseases. Myeloid-specific deletion of miR-182 protects mice against excessive osteoclastogenesis and bone resorption in disease models of ovariectomy-induced osteoporosis and inflammatory arthritis. Pharmacological treatment of these diseases with miR-182 inhibitors completely suppresses pathologic bone erosion. Mechanistically, we identify protein kinase double-stranded RNA-dependent (PKR) as a new and essential miR-182 target that is a novel inhibitor of osteoclastogenesis via regulation of the endogenous interferon (IFN)-β-mediated autocrine feedback loop. The expression levels of miR-182, PKR, and IFN-β are altered in RA and are significantly correlated with the osteoclastogenic capacity of RA monocytes. Our findings reveal a previously unrecognized regulatory network mediated by miR-182-PKR-IFN-β axis in osteoclastogenesis, and highlight the therapeutic implications of miR-182 inhibition in osteoprotection. Osteoclasts mediate bone disruption in a number of degenerative bone diseases. Here, the authors show that miR-182 regulates osteoclastogenesis via PKR and IFN-beta signaling, is correlated with rheumatoid arthritis, and that its ablation or inhibition is protective against bone erosion in mouse models of osteoporosis or inflammatory arthritis.
Collapse
|
31
|
Transcriptome changes in muscle of Nellore cows submitted to recovery weight gain under grazing condition. Animal 2018; 13:333-340. [PMID: 29983126 DOI: 10.1017/s1751731118001490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to evaluate transcriptome changes in the muscle tissue of Bos taurus indicus cull cows subjected to recovery weight gain under grazing conditions. In all, 38 Nellore cull cows were divided randomly into two different management groups: (1) Maintenance (MA) and (2) Recovery gain (RG) from weight loss by moderate growth under high forage availability. After slaughter, RNA analysis was performed on the Longissimus thoracis muscle. Semaphorin 4A, solute carrier family 11 member 1, and Ficolin-2 were expressed in the RG, which may indicate an inflammatory response during tissue regrowth. Signaling factors, such as Myostatin, related to fibroblast activation, negative control of satellite cell proliferation in adults and muscle protein synthesis were less abundant in the RG group. The only gene related to anabolic processes that were more abundant in the MA group was related to fat deposition. The genes that were differentially expressed in the experiment showed muscle repair-related changes during RG based on the greater expression of genes involved in inflammatory responses and the lower expression of negative regulators of muscle cell proliferation and hypertrophy.
Collapse
|
32
|
Liu Y, Yue P, Zhou T, Zhang F, Wang H, Chen X. LncRNA MEG3 enhances 131I sensitivity in thyroid carcinoma via sponging miR-182. Biomed Pharmacother 2018; 105:1232-1239. [PMID: 30021359 DOI: 10.1016/j.biopha.2018.06.087] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/14/2018] [Accepted: 06/14/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Long non-coding RNA (LncRNA) MEG3 has been demonstrated as a tumor suppressor in various cancers, including thyroid carcinoma (TC). However, the detail functions and possible mechanisms of MEG3 in 131I resistance of TC remain to be uncovered. METHODS qRT-PCR was performed for the detection of MEG3 and miR-182 levels. 131I-resistant TC cells were constructed by continuous exposure to stepwise increased concentrations of 131I. Western blot assay was used to measure the protein expressions of γ-H2 AX and H2 AX. CCK-8 and flow cytometry assays were carried out for the evaluation of cell viability and apoptosis, respectively. Bioinformatics and dual-luciferse assays were conducted to prove the interaction of MEG3 and miR-182. RESULTS MEG3 expression was down-regulated in TC tumor tissues, and the cumulative survival rate was decreased in low MEG3 expression group in TC patients under 131I treatment. MEG3 expression appeared a decline and miR-182 expression displayed an increase in 131I-resistant FTC-133 (res-FTC-133) and TPC-1 (res-TPC-1) cells. Moreover, MEG3 overexpression suppressed 131I-resistant cell viability, promoted apoptosis and induced DNA damage. MEG3 was verified as a molecular sponge for miR-182, and inhibition of miR-182 exerted similar functions as MEG3 overexpression. Furthermore, MEG3 knockdown substantially abrogated the anti-cancer functions of anti-miR-182. CONCLUSIONS MEG3 enhanced the radiosensitivity of 131I in TC cells via sponging miR-182, indicating that MEG3 may act as a potential biomarker and therapeutic target for TC patients with 131I resistance.
Collapse
Affiliation(s)
- Yang Liu
- Department of Nuclear Medicine, The First People's Hospital of Shangqiu, Shangqiu, 476000, China
| | - Peiru Yue
- Department of Oncology, The First People's Hospital of Shangqiu, Shangqiu, 476000, China
| | - Tao Zhou
- Department of Nuclear Medicine, The First People's Hospital of Shangqiu, Shangqiu, 476000, China
| | - Fengzhen Zhang
- Department of Nuclear Medicine, The First People's Hospital of Shangqiu, Shangqiu, 476000, China
| | - Huixiang Wang
- Department of Nuclear Medicine, The First People's Hospital of Shangqiu, Shangqiu, 476000, China
| | - Xiaoqi Chen
- Department of Digestive Oncology, the First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China.
| |
Collapse
|
33
|
Liu X, Li H, Wu G, Cui S. miR-182 promotes cell proliferation and invasion by inhibiting APC in melanoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:1900-1908. [PMID: 31938296 PMCID: PMC6958208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/14/2018] [Indexed: 06/10/2023]
Abstract
OBJECTIVE Common treatment methods have shown a lack of therapeutic effect in melanoma, a type of malignant tumor. The pathogenesis of melanoma is not yet fully clear, therefore, search for a new treatment strategy is urgent. Recent studies have demonstrated that miR-182 is remarkably over-expressed in human melanoma. Our study aimed to explore the underlying mechanism of miR-182 on melanoma. METHODS In this study, the expression level of miR-182 was detected using RT-PCR in melanoma and adjacent tissues as well as in HEM-m, A375, A2058, and WM35 cell lines. Functions of miR-182 were investigated by using CCK-8 on cell proliferation, apoptosis, invasion, and cell cycle on A375 cells. Moreover, expression levels of Frz, Dsh, β-catenin, APC, Axin, GSK-3β, and CK1 were detected by Western blotting after knockdown and overexpression of miR-182. Overexpression of miR-182 and knockdown of APC was used to demonstrate regulated functions in melanoma. RESULTS Expression levels of miR-182 were significantly upregulated in melanoma tissues and cell lines. Overexpression of miR-182 promoted cell proliferation, migration, and invasion while inhibiting cell apoptosis and cell cycle in S phase. Overexpression of miR-182 upregulated expression levels of β-catenin and APC. Overexpression of miR-182 and knockdown of APC inhibited proliferation of melanoma cells and tumors. CONCLUSION Overexpression of miR-182 promotes cell proliferation and invasion by targeting to APC in melanoma. The pathogenesis of miR-182 and APC might provide therapeutic targets for treatment of melanoma in the molecular level.
Collapse
Affiliation(s)
- Xilin Liu
- Department of Hand Surgery, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin Province, China
| | - Hong Li
- Department of Rehabilitation, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin Province, China
| | - Guangzhi Wu
- Department of Hand Surgery, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin Province, China
| | - Shusen Cui
- Department of Hand Surgery, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin Province, China
| |
Collapse
|
34
|
Abstract
MicroRNAs (miRNAs) are ∼22 nt RNAs that direct posttranscriptional repression of mRNA targets in diverse eukaryotic lineages. In humans and other mammals, these small RNAs help sculpt the expression of most mRNAs. This article reviews advances in our understanding of the defining features of metazoan miRNAs and their biogenesis, genomics, and evolution. It then reviews how metazoan miRNAs are regulated, how they recognize and cause repression of their targets, and the biological functions of this repression, with a compilation of knockout phenotypes that shows that important biological functions have been identified for most of the broadly conserved miRNAs of mammals.
Collapse
Affiliation(s)
- David P Bartel
- Howard Hughes Medical Institute and Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
35
|
Ye YY, Mei JW, Xiang SS, Li HF, Ma Q, Song XL, Wang Z, Zhang YC, Liu YC, Jin YP, Hu YP, Jiang L, Liu FT, Zhang YJ, Hao YJ, Liu YB. MicroRNA-30a-5p inhibits gallbladder cancer cell proliferation, migration and metastasis by targeting E2F7. Cell Death Dis 2018; 9:410. [PMID: 29540696 PMCID: PMC5852001 DOI: 10.1038/s41419-018-0444-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/07/2018] [Accepted: 02/15/2018] [Indexed: 12/12/2022]
Abstract
Gallbladder carcinoma (GBC), the most common malignant tumour of the bile duct, is highly aggressive and has a poor prognosis. MicroRNA-30a-5p (miR-30a-5p) is an important tumour suppressor that participates in many aspects of carcinogenesis and cancer development. However, the role of miR-30a-5p in GBC development remains to be determined, as do the mechanisms underlying its effects in GBC. Using samples collected from 42 subjects with gallbladder carcinoma (GBC), we showed decreased miR-30a-5p expression in the primary lesions vs. non-tumour adjacent tissues (NATs). Decreased miR-30a-5p was associated with shorter disease-free survival (DFS) and overall survival (OS). Inhibiting miR-30a-5p expression in 2 representative GBC cell lines (GBC-SD and NOZ) increased cell proliferation, migration, invasiveness, as well as β-catenin nuclear translocation, vice versa. In nude mice, NOZ cells transfected with miR-30a-5p mimics grew slower (vs. miR-NC) upon subcutaneous inoculation, and had lower rate of hepatic metastasis upon spleen inoculation. Dual luciferase assay confirmed that E2F transcription factor 7 (E2F7) was a direct target of miR-30a-5p and antagonized the effects induced by miR-30a-5p downregulation in GBC cells. MiR-30a-5p attenuates the EMT and metastasis in GBC cells by targeting E2F7, suggesting miR-30a-5p is a tumour suppressor that may serve as a novel potential prognostic biomarker or molecular therapeutic target for GBC.
Collapse
Affiliation(s)
- Yuan-Yuan Ye
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Jia-Wei Mei
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Shan-Shan Xiang
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Huai-Feng Li
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Qiang Ma
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Xiao-Ling Song
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Zheng Wang
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yi-Chi Zhang
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yong-Chen Liu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yun-Peng Jin
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yun-Ping Hu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Lin Jiang
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Fa-Tao Liu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yi-Jian Zhang
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Ya-Juan Hao
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China.
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China.
| | - Ying-Bin Liu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
- Shanghai Key Laboratory of Biliary Tract Disease Research, 1665 Kongjiang Road, Shanghai, 200092, China.
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
36
|
Segura MF, Jubierre L, Li S, Soriano A, Koetz L, Gaziel-Sovran A, Masanas M, Kleffman K, Dankert JF, Walsh MJ, Hernando E. Krüppel-like factor 4 (KLF4) regulates the miR-183~96~182 cluster under physiologic and pathologic conditions. Oncotarget 2018; 8:26298-26311. [PMID: 28412746 PMCID: PMC5432258 DOI: 10.18632/oncotarget.15459] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 02/06/2017] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of endogenous non-coding small RNAs that post-transcriptionally control the translation and stability of target mRNAs in a sequence-dependent manner. MiRNAs are essential for key cellular processes including proliferation, differentiation, cell death and metabolism, among others. Consequently, alterations of miRNA expression contribute to developmental defects and a myriad of diseases.The expression of miRNAs can be altered by several mechanisms including gene copy number alterations, aberrant DNA methylation, defects of the miRNA processing machinery or unscheduled expression of transcription factors. In this work, we sought to analyze the regulation of the miR-182 cluster, located at the 7q32 locus, which encodes three different miRNAs that are abundantly expressed in human embryonic stem cells and de-regulated in cancer. We have found that the Krüppel-like factor 4 (KLF4) directly regulates miR-182 cluster expression in human embryonic stem cells (hESCs) and in melanoma tumors, in which the miR-182 cluster is highly expressed and has a pro-metastatic role. Furthermore, higher KLF4 expression was found to be associated with metastatic progression and poor patient outcome. Loss of function experiments revealed that KLF4 is required for melanoma cell maintenance. These findings provide new insights into the regulation of the miR-182 cluster expression and new opportunities for therapeutic intervention in tumors in which the KLF4-miR-182 cluster axis is deregulated.
Collapse
Affiliation(s)
- Miguel F Segura
- Department of Pathology, New York University School of Medicine, New York, NY, USA.,Interdisciplinary Melanoma Cooperative Group, New York University Perlmutter Cancer Institute, NYU School of Medicine, New York, NY, USA.,Laboratory of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute (VHIR)-UAB, Barcelona, Spain
| | - Luz Jubierre
- Laboratory of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute (VHIR)-UAB, Barcelona, Spain
| | - SiDe Li
- Departments of Structural and Chemical Biology, Genetics and Genomic Sciences and Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aroa Soriano
- Laboratory of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute (VHIR)-UAB, Barcelona, Spain
| | - Lisa Koetz
- Department of Pathology, New York University School of Medicine, New York, NY, USA.,Interdisciplinary Melanoma Cooperative Group, New York University Perlmutter Cancer Institute, NYU School of Medicine, New York, NY, USA
| | - Avital Gaziel-Sovran
- Department of Pathology, New York University School of Medicine, New York, NY, USA.,Interdisciplinary Melanoma Cooperative Group, New York University Perlmutter Cancer Institute, NYU School of Medicine, New York, NY, USA
| | - Marc Masanas
- Laboratory of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute (VHIR)-UAB, Barcelona, Spain
| | - Kevin Kleffman
- Department of Pathology, New York University School of Medicine, New York, NY, USA.,Interdisciplinary Melanoma Cooperative Group, New York University Perlmutter Cancer Institute, NYU School of Medicine, New York, NY, USA
| | - John F Dankert
- Department of Pathology, New York University School of Medicine, New York, NY, USA.,Interdisciplinary Melanoma Cooperative Group, New York University Perlmutter Cancer Institute, NYU School of Medicine, New York, NY, USA
| | - Martin J Walsh
- Departments of Structural and Chemical Biology, Genetics and Genomic Sciences and Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eva Hernando
- Department of Pathology, New York University School of Medicine, New York, NY, USA.,Interdisciplinary Melanoma Cooperative Group, New York University Perlmutter Cancer Institute, NYU School of Medicine, New York, NY, USA
| |
Collapse
|
37
|
Ma Y, Liang AJ, Fan YP, Huang YR, Zhao XM, Sun Y, Chen XF. Dysregulation and functional roles of miR-183-96-182 cluster in cancer cell proliferation, invasion and metastasis. Oncotarget 2018; 7:42805-42825. [PMID: 27081087 PMCID: PMC5173173 DOI: 10.18632/oncotarget.8715] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/31/2016] [Indexed: 02/07/2023] Open
Abstract
Previous studies have reported aberrant expression of the miR-183-96-182 cluster in a variety of tumors, which indicates its' diagnostic or prognostic value. However, a key characteristic of the miR-183-96-182 cluster is its varied expression levels, and pleomorphic functional roles in different tumors or under different conditions. In most tumor types, the cluster is highly expressed and promotes tumorigenesis, cancer progression and metastasis; yet tumor suppressive effects have also been reported in some tumors. In the present study, we discuss the upstream regulators and the downstream target genes of miR-183-96-182 cluster, and highlight the dysregulation and functional roles of this cluster in various tumor cells. Newer insights summarized in this review will help readers understand the different facets of the miR-183-96-182 cluster in cancer development and progression.
Collapse
Affiliation(s)
- Yi Ma
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - A-Juan Liang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yu-Ping Fan
- Reproductive Medicine Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi-Ran Huang
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Ming Zhao
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yun Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Xiang-Feng Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China.,Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
38
|
Jia L, Luo S, Ren X, Li Y, Hu J, Liu B, Zhao L, Shan Y, Zhou H. miR-182 and miR-135b Mediate the Tumorigenesis and Invasiveness of Colorectal Cancer Cells via Targeting ST6GALNAC2 and PI3K/AKT Pathway. Dig Dis Sci 2017; 62:3447-3459. [PMID: 29030743 DOI: 10.1007/s10620-017-4755-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 09/07/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND Metastasis is a leading cause of cancer-related death including colorectal cancer (CRC). MicroRNAs are known to regulate cancer pathways and to be expressed aberrantly in cancer. Aberrant sialylation is closely associated with malignant phenotype of tumor cells, including invasiveness and metastasis. AIM This study aimed to investigate the association of miR-182 and miR-135b with proliferation and invasion by targeting sialyltransferase ST6GALNAC2 in CRC cells and explore the potential molecular mechanism. METHODS We measured the levels of miR-182, miR-135b, and ST6GALNAC2 in a series of CRC cell lines and tissues using real-time PCR. Bioinformatics analysis and luciferase reporter assay were performed to test the direct binding of miR-182 and miR-135b to the target gene ST6GALNAC2. We also analyzed the possible role of miR-182/-135b on colony formation, wound healing, invasion, and tube formation. RESULTS The expression of miR-182 and miR-135b was higher in tumor tissues compared to adjacent noncancerous tissues of CRC patients, as well as up-regulated in SW620 cells than in SW480 cells with different metastatic potential. By applying bioinformatics analysis and luciferase reporter assay, we identified ST6GALNAC2 as the direct target of miR-182/-135b. Furthermore, miR-182/-135b inhibited significantly ST6GALNAC2 expression, and consistently, ST6GALNAC2 mediated migration, adhesion, invasion, proliferation, and tumor angiogenesis in CRC cell lines. Additionally, PI3K/AKT signaling pathway was regulated by miR-182/135b, which was partially blocked by altered level of ST6GALNAC2 in CRC. CONCLUSIONS The miR-182/-135b/ST6GALNAC2/PI3K/AKT axis may serve as a predictive biomarker and a potential therapeutic target in CRC treatment.
Collapse
Affiliation(s)
- Li Jia
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China.
| | - Shihua Luo
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
- Department of Traumatology, Shanghai Ruijin Hospital, Jiaotong University, Shanghai, 200025, China
| | - Xiang Ren
- College of Stomatology, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Yang Li
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Jialei Hu
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Bing Liu
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Lifen Zhao
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Yujia Shan
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Huimin Zhou
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| |
Collapse
|
39
|
Stereotactic Body Radiotherapy for Oligometastasis: Opportunities for Biology to Guide Clinical Management. Cancer J 2017; 22:247-56. [PMID: 27441744 DOI: 10.1097/ppo.0000000000000202] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Oligometastasis refers to a state of limited metastatic disease burden, in which surgical or ablative treatment to all known visible metastases holds promise to extend survival or even effect cure. Stereotactic body radiotherapy is a form of radiation treatment capable of delivering a high biologically effective dose of radiation in a highly conformal manner, with a favorable toxicity profile. Enthusiasm for oligometastasis ablation, however, should be counterbalanced against the limited supporting evidence. It remains unknown to what extent (if any) ablation influences survival or quality of life. Rising clinical equipoise necessitates the completion of randomized controlled trials to assess this, several of which are underway. However, a lack of clear identification criteria or biomarkers to define the oligometastatic state hampers optimal patient selection.This narrative review explores the evolutionary origins of oligometastasis, the steps of the metastatic process at which oligometastases may arise, and the biomolecular mediators of this state. It discusses clinical outcomes with treatment of oligometastases, ongoing trials, and areas of basic and translational research that may lead to novel biomarkers. These efforts should provide a clearer, biomolecular definition of oligometastatic disease and aid in the accurate selection of patients for ablative therapies.
Collapse
|
40
|
Dodd RD, Lee CL, Overton T, Huang W, Eward WC, Luo L, Ma Y, Ingram DR, Torres KE, Cardona DM, Lazar AJ, Kirsch DG. NF1 +/- Hematopoietic Cells Accelerate Malignant Peripheral Nerve Sheath Tumor Development without Altering Chemotherapy Response. Cancer Res 2017; 77:4486-4497. [PMID: 28646022 PMCID: PMC5839126 DOI: 10.1158/0008-5472.can-16-2643] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 03/09/2017] [Accepted: 06/12/2017] [Indexed: 12/11/2022]
Abstract
Haploinsufficiency in the tumor suppressor NF1 contributes to the pathobiology of neurofibromatosis type 1, but a related role has not been established in malignant peripheral nerve sheath tumors (MPNST) where NF1 mutations also occur. Patients with NF1-associated MPNST appear to have worse outcomes than patients with sporadic MPNST, but the mechanism underlying this correlation is not understood. To define the impact of stromal genetics on the biology of this malignancy, we developed unique mouse models that reflect the genetics of patient-associated MPNST. Specifically, we used adenovirus-Cre injections to generate MPNST in Nf1Flox/Flox; Ink4a/ArfFlox/Flox and Nf1Flox/-; Ink4a/ArfFlox/Flox paired littermate mice to model tumors from NF1-wild-type and NF1-associated patients, respectively. In these models, Nf1 haploinsufficiency in hematopoietic cells accelerated tumor onset and increased levels of tumor-infiltrating immune cells comprised of CD11b+ cells, monocytes, and mast cells. We observed that mast cells were also enriched in human NF1-associated MPNST. In a coclinical trial to examine how the tumor microenvironment influences the response to multiagent chemotherapy, we found that stromal Nf1 status had no effect. Taken together, our results clarify the role of the NF1-haploinsufficient tumor microenvironment in MPNST. Cancer Res; 77(16); 4486-97. ©2017 AACR.
Collapse
Affiliation(s)
- Rebecca D Dodd
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Chang-Lung Lee
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Tess Overton
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Wesley Huang
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - William C Eward
- Department of Orthopaedic Surgery, Duke University, Durham, North Carolina
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Davis R Ingram
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Keila E Torres
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Diana M Cardona
- Department of Pathology, Duke University, Durham, North Carolina
| | - Alexander J Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David G Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina.
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
41
|
Huang J, Chen M, Whitley MJ, Kuo HC, Xu ES, Walens A, Mowery YM, Van Mater D, Eward WC, Cardona DM, Luo L, Ma Y, Lopez OM, Nelson CE, Robinson-Hamm JN, Reddy A, Dave SS, Gersbach CA, Dodd RD, Kirsch DG. Generation and comparison of CRISPR-Cas9 and Cre-mediated genetically engineered mouse models of sarcoma. Nat Commun 2017; 8:15999. [PMID: 28691711 PMCID: PMC5508130 DOI: 10.1038/ncomms15999] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 05/17/2017] [Indexed: 01/03/2023] Open
Abstract
Genetically engineered mouse models that employ site-specific recombinase technology are important tools for cancer research but can be costly and time-consuming. The CRISPR-Cas9 system has been adapted to generate autochthonous tumours in mice, but how these tumours compare to tumours generated by conventional recombinase technology remains to be fully explored. Here we use CRISPR-Cas9 to generate multiple subtypes of primary sarcomas efficiently in wild type and genetically engineered mice. These data demonstrate that CRISPR-Cas9 can be used to generate multiple subtypes of soft tissue sarcomas in mice. Primary sarcomas generated with CRISPR-Cas9 and Cre recombinase technology had similar histology, growth kinetics, copy number variation and mutational load as assessed by whole exome sequencing. These results show that sarcomas generated with CRISPR-Cas9 technology are similar to sarcomas generated with conventional modelling techniques and suggest that CRISPR-Cas9 can be used to more rapidly generate genotypically and phenotypically similar cancers.
Collapse
Affiliation(s)
- Jianguo Huang
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Mark Chen
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
- Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Melodi Javid Whitley
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
- Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Hsuan-Cheng Kuo
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Eric S. Xu
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Andrea Walens
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Yvonne M. Mowery
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - David Van Mater
- Division of Hematology-Oncology, Department of Pediatrics, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - William C. Eward
- Department of Orthopedic Surgery, Duke University, Durham, North Carolina 27710, USA
| | - Diana M. Cardona
- Department of Pathology, Duke University, Durham, North Carolina 27710, USA
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Omar M. Lopez
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Christopher E. Nelson
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
- Duke Center for Genomic and Computational Biology, Duke University, Durham, North Carolina 27708, USA
| | - Jacqueline N. Robinson-Hamm
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
- Duke Center for Genomic and Computational Biology, Duke University, Durham, North Carolina 27708, USA
| | - Anupama Reddy
- Duke Center for Genomic and Computational Biology, Duke University, Durham, North Carolina 27708, USA
| | - Sandeep S. Dave
- Duke Center for Genomic and Computational Biology, Duke University, Durham, North Carolina 27708, USA
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Charles A. Gersbach
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
- Duke Center for Genomic and Computational Biology, Duke University, Durham, North Carolina 27708, USA
| | - Rebecca D. Dodd
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - David G. Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
42
|
Wurm AA, Zjablovskaja P, Kardosova M, Gerloff D, Bräuer-Hartmann D, Katzerke C, Hartmann JU, Benoukraf T, Fricke S, Hilger N, Müller AM, Bill M, Schwind S, Tenen DG, Niederwieser D, Alberich-Jorda M, Behre G. Disruption of the C/EBPα-miR-182 balance impairs granulocytic differentiation. Nat Commun 2017; 8:46. [PMID: 28663557 PMCID: PMC5491528 DOI: 10.1038/s41467-017-00032-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 04/30/2017] [Indexed: 02/04/2023] Open
Abstract
Transcription factor C/EBPα is a master regulator of myelopoiesis and its inactivation is associated with acute myeloid leukemia. Deregulation of C/EBPα by microRNAs during granulopoiesis or acute myeloid leukemia development has not been studied. Here we show that oncogenic miR-182 is a strong regulator of C/EBPα. Moreover, we identify a regulatory loop between C/EBPα and miR-182. While C/EBPα blocks miR-182 expression by direct promoter binding during myeloid differentiation, enforced expression of miR-182 reduces C/EBPα protein level and impairs granulopoiesis in vitro and in vivo. In addition, miR-182 expression is highly elevated particularly in acute myeloid leukemia patients with C-terminal CEBPA mutations, thereby depicting a mechanism by which C/EBPα blocks miR-182 expression. Furthermore, we present miR-182 expression as a prognostic marker in cytogenetically high-risk acute myeloid leukemia patients. Our data demonstrate the importance of a controlled balance between C/EBPα and miR-182 for the maintenance of healthy granulopoiesis. C/EBPα is a critical transcription factor involved in myelopoiesis and its inactivation is associated with acute myeloid leukemia (AML). Here the authors show a negative feedback loop between C/EBPα and miR-182 and identify this miRNA as a marker of high-risk AML.
Collapse
Affiliation(s)
- Alexander Arthur Wurm
- Division of Hematology and Oncology, Leipzig University Hospital, Johannisallee 32a, Leipzig, 04103, Germany
| | - Polina Zjablovskaja
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Videnska 1083, Prague 4, 142 20, Czech Republic
| | - Miroslava Kardosova
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Videnska 1083, Prague 4, 142 20, Czech Republic
| | - Dennis Gerloff
- Division of Hematology and Oncology, Leipzig University Hospital, Johannisallee 32a, Leipzig, 04103, Germany
| | - Daniela Bräuer-Hartmann
- Division of Hematology and Oncology, Leipzig University Hospital, Johannisallee 32a, Leipzig, 04103, Germany
| | - Christiane Katzerke
- Division of Hematology and Oncology, Leipzig University Hospital, Johannisallee 32a, Leipzig, 04103, Germany
| | - Jens-Uwe Hartmann
- Division of Hematology and Oncology, Leipzig University Hospital, Johannisallee 32a, Leipzig, 04103, Germany
| | - Touati Benoukraf
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore
| | - Stephan Fricke
- Fraunhofer Institute for Cell Therapy and Immunology, Perlickstraße 1, Leipzig, 04103, Germany
| | - Nadja Hilger
- Fraunhofer Institute for Cell Therapy and Immunology, Perlickstraße 1, Leipzig, 04103, Germany
| | - Anne-Marie Müller
- Fraunhofer Institute for Cell Therapy and Immunology, Perlickstraße 1, Leipzig, 04103, Germany
| | - Marius Bill
- Division of Hematology and Oncology, Leipzig University Hospital, Johannisallee 32a, Leipzig, 04103, Germany
| | - Sebastian Schwind
- Division of Hematology and Oncology, Leipzig University Hospital, Johannisallee 32a, Leipzig, 04103, Germany
| | - Daniel G Tenen
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore.,Harvard Stem Cell Institute, Harvard Medical School, 3 Blackfan Circle, Boston, MA, 02115, USA
| | - Dietger Niederwieser
- Division of Hematology and Oncology, Leipzig University Hospital, Johannisallee 32a, Leipzig, 04103, Germany
| | - Meritxell Alberich-Jorda
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Videnska 1083, Prague 4, 142 20, Czech Republic
| | - Gerhard Behre
- Division of Hematology and Oncology, Leipzig University Hospital, Johannisallee 32a, Leipzig, 04103, Germany.
| |
Collapse
|
43
|
Chen L, Zhang C, Wang Y, Li Y, Han Q, Yang H, Zhu Y. Data mining and pathway analysis of glucose-6-phosphate dehydrogenase with natural language processing. Mol Med Rep 2017. [PMID: 28627690 PMCID: PMC5562079 DOI: 10.3892/mmr.2017.6785] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Human glucose-6-phosphate dehydrogenase (G6PD) is a crucial enzyme in the pentose phosphate pathway, and serves an important role in biosynthesis and the redox balance. G6PD deficiency is a major cause of neonatal jaundice and acute hemolyticanemia, and recently, G6PD has been associated with diseases including inflammation and cancer. The aim of the present study was to conduct a search of the National Center for Biotechnology Information PubMed library for articles discussing G6PD. Genes that were identified to be associated with G6PD were recorded, and the frequency at which each gene appeared was calculated. Gene ontology (GO), pathway and network analyses were then performed. A total of 98 G6PD‑associated genes and 33 microRNAs (miRNAs) that potentially regulate G6PD were identified. The 98 G6PD‑associated genes were then sub‑classified into three functional groups by GO analysis, followed by analysis of function, pathway, network, and disease association. Out of the 47 signaling pathways identified, seven were significantly correlated with G6PD‑associated genes. At least two out of four independent programs identified the 33 miRNAs that were predicted to target G6PD. miR‑1207‑5P, miR‑1 and miR‑125a‑5p were predicted by all four software programs to target G6PD. The results of the present study revealed that dysregulation of G6PD was associated with cancer, autoimmune diseases, and oxidative stress‑induced disorders. These results revealed the potential roles of G6PD‑regulated signaling and metabolic pathways in the etiology of these diseases.
Collapse
Affiliation(s)
- Long Chen
- Department of Biochemistry and Molecular Biology of Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Chunhua Zhang
- Department of Biochemistry and Molecular Biology of Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yanling Wang
- Department of Biochemistry and Molecular Biology of Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yuqian Li
- Department of Biochemistry and Molecular Biology of Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Qiaoqiao Han
- Department of Biochemistry and Molecular Biology of Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Huixin Yang
- Department of Biochemistry and Molecular Biology of Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yuechun Zhu
- Department of Biochemistry and Molecular Biology of Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
44
|
Grisard E, Nicoloso MS. Following MicroRNAs Through the Cancer Metastatic Cascade. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 333:173-228. [PMID: 28729025 DOI: 10.1016/bs.ircmb.2017.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Approximately a decade ago the first MicroRNAs (MiRNAs) participating in cancer metastasis were identified and metastmiRs were initially only a handful. Since those first reports, MiRNA research has explosively thrived, mainly due to their revolutionary mechanism of action and the hope of having at hand a novel tool to control cancer aggressiveness. This has ultimately led to delineate an almost impenetrable regulatory network: hundreds of MiRNAs transversally dominating every aspect of normal and cancer biology, each MiRNA having hundreds of targets and context-dependent activity. Providing a comprehensive description of MiRNA roles in cancer metastasis is a daunting task; nevertheless, we still believe that grasping the big picture of MiRNAs in cancer metastasis can give a different perspective on the potential insights and approaches that MiRNAs can offer to understand cancer complexity (e.g., as predictive and prognostic markers) and to tackle cancer metastasis (e.g., as therapeutic targets or tools). This chapter presents a schematic overview of the role of MiRNAs in governing cancer metastasis, describing step by step the cellular and molecular processes whereby cancer cells conquer distant organs and can grow as secondary tumors at different distant sites, and for each step, we will introduce how MiRNAs impinge on each one of them. We deeply apologize with our colleagues for any of their research work that, for clarity, for our effort to streamline and due to space limitations, we did not cite.
Collapse
|
45
|
Urbánek P, Klotz L. Posttranscriptional regulation of FOXO expression: microRNAs and beyond. Br J Pharmacol 2017; 174:1514-1532. [PMID: 26920226 PMCID: PMC5446586 DOI: 10.1111/bph.13471] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 02/18/2016] [Accepted: 02/23/2016] [Indexed: 01/17/2023] Open
Abstract
Forkhead box, class O (FOXO) transcription factors are major regulators of diverse cellular processes, including fuel metabolism, oxidative stress response and redox signalling, cell cycle progression and apoptosis. Their activities are controlled by multiple posttranslational modifications and nuclear-cytoplasmic shuttling. Recently, post-transcriptional regulation of FOXO synthesis has emerged as a new regulatory level of their functions. Accumulating evidence suggests that this post-transcriptional mode of regulation of FOXO activity operates in response to stressful stimuli, including oxidative stress. Here, we give a brief overview on post-transcriptional regulation of FOXO synthesis by microRNAs (miRNAs) and by RNA-binding regulatory proteins, human antigen R (HuR) and quaking (QKI). Aberrant post-transcriptional regulation of FOXOs is frequently connected with various disease states. We therefore discuss characteristic examples of FOXO regulation at the post-transcriptional level under various physiological and pathophysiological conditions, including oxidative stress and cancer. The picture emerging from this summary points to a diversity of interactions between miRNAs/miRNA-induced silencing complexes and RNA-binding regulatory proteins. Better insight into these complexities of post-transcriptional regulatory interactions will add to our understanding of the mechanisms of pathological processes and the role of FOXO proteins. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- P Urbánek
- Institute of Nutrition, Department of NutrigenomicsFriedrich‐Schiller‐Universität JenaJenaGermany
| | - L‐O Klotz
- Institute of Nutrition, Department of NutrigenomicsFriedrich‐Schiller‐Universität JenaJenaGermany
| |
Collapse
|
46
|
Samantarrai D, Mallick B. miR-429 inhibits metastasis by targeting KIAA0101 in Soft Tissue Sarcoma. Exp Cell Res 2017; 357:33-39. [PMID: 28432002 DOI: 10.1016/j.yexcr.2017.04.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 04/11/2017] [Accepted: 04/17/2017] [Indexed: 12/17/2022]
Abstract
Soft tissue sarcomas (STS) are a heterogeneous group of rare tumors with high metastatic potential. There being only a handful of publication on metastasis of STS, we investigated the miRNA mediated target gene regulations in modulating the metastatic processes in this cancer. In this study, we amalgamated gene and miRNA expression profiles of high-grade STS samples with miRNA target predictions and identified miR-429 targeting KIAA0101 as a novel pair, which remain unexplored in STS metastasis. We validated their expression in metastatic fibrosarcoma cell line, HT1080 and performed several functional assays using miRNA mimics and KIAA0101 over-expression vector to confirm their role in metastasis. We observed miR-429 is downregulated in HT1080 cells and acting as an anti-metastatic miRNA that inhibited proliferation, migration, anchorage independent growth and invasion by de-repressing KIAA0101. Moreover, the renilla luciferase reporter assay confirmed that miR-429 targets KIAA0101 by binding to its 3/UTR and influence its expression. Taken together, our work demonstrated miR-429 mediates deregulation of KIAA0101 by acting as an anti-metastatic miRNA that targets KIAA0101 pro-metastatic gene during metastasis of STS.
Collapse
Affiliation(s)
- Devyani Samantarrai
- RNAi and Functional Genomics Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha 769008, India
| | - Bibekanand Mallick
- RNAi and Functional Genomics Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha 769008, India.
| |
Collapse
|
47
|
López-Urrutia E, Coronel-Hernández J, García-Castillo V, Contreras-Romero C, Martínez-Gutierrez A, Estrada-Galicia D, Terrazas LI, López-Camarillo C, Maldonado-Martínez H, Jacobo-Herrera N, Pérez-Plasencia C. MiR-26a downregulates retinoblastoma in colorectal cancer. Tumour Biol 2017; 39:1010428317695945. [PMID: 28443472 DOI: 10.1177/1010428317695945] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs are non-coding short RNAs that target the 3' untranslated region of messenger RNAs (mRNAs) and lead to their degradation or to translational repression. Several microRNAs have been designated as oncomirs, owing to their regulating tumor suppressor genes. Interestingly, a few of them have been found to target multiple genes whose simultaneous suppression contributes to the development of a tumoral phenotype. Here, we have showed that miR-26a is overexpressed in colorectal cancer data obtained from TCGA Research Network and in human colon cancer pathological specimens; moreover, an orthotopic in vivo model of colon cancer showed overexpression of miR-26a, while Rb1 expression inversely correlated to miR-26a in TCGA Research Network data, pathological samples, and the in vivo model. Then, by means of luciferase assay, we demonstrated that miR-26a targets the 3' untranslated region of Rb1 mRNA directly. This is, to our knowledge, the first report of miR-26a targeting Rb1 in colon cancer. The results of this study suggested that miR-26a could serve as a progression biomarker in colorectal cancer. Further validation studies are still needed to confirm our findings.
Collapse
Affiliation(s)
- Eduardo López-Urrutia
- 1 Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, México
| | - Jossimar Coronel-Hernández
- 1 Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, México
| | - Verónica García-Castillo
- 1 Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, México
| | - Carlos Contreras-Romero
- 1 Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, México
| | - Antonio Martínez-Gutierrez
- 1 Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, México
| | - Diana Estrada-Galicia
- 1 Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, México
| | - Luis Ignacio Terrazas
- 2 Laboratorio de Inmunología, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, Mexico
| | - César López-Camarillo
- 3 Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico City, Mexico
| | | | - Nadia Jacobo-Herrera
- 5 Unidad de Bioquímica, Instituto Nacional De Ciencias Médicas Y Nutrición Salvador Zubirán, Tlalpan, Mexico
| | - Carlos Pérez-Plasencia
- 1 Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, México
- 6 Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, México
| |
Collapse
|
48
|
Gandellini P, Doldi V, Zaffaroni N. microRNAs as players and signals in the metastatic cascade: Implications for the development of novel anti-metastatic therapies. Semin Cancer Biol 2017; 44:132-140. [PMID: 28344166 DOI: 10.1016/j.semcancer.2017.03.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/21/2017] [Accepted: 03/21/2017] [Indexed: 01/28/2023]
Abstract
microRNAs (miRNAs) are small non-coding RNAs that negatively regulate gene expression at the post-transcriptional level. Increasing evidence emerging from human tumor preclinical models clearly indicates that specific miRNAs, collectively termed "metastamirs," play a functional role in different steps of the metastatic cascade, by exerting either pro- or anti-metastatic functions, and behave as signaling mediators to enable tumor cell to colonize a specific organ. miRNAs also actively participate in the proficient interaction of cancer cells with tumor microenvironment, either at the primary or at the metastatic site. Circulating miRNAs, released by multiple cell types, following binding to proteins or encapsulation in extracellular vesicles, play a main role in this cross-talk by acting as transferrable messages. The documented involvement of specific miRNAs in the dissemination process has aroused interest in the development of miRNA-based strategies for the treatment of metastasis. Preclinical research carried out in tumor experimental models, using both miRNA replacement and miRNA inhibitory approaches, is encouraging towards translating miRNA-based strategies into human cancer therapy, based on the observed therapeutic activity in the absence of main toxicity. However, to accelerate their adoption in the clinic, further improvements in terms of efficacy and targeted delivery to the tumor are still necessary.
Collapse
Affiliation(s)
- Paolo Gandellini
- Molecular Pharmacology, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Via Amadeo 42, 20133 Milan, Italy.
| | - Valentina Doldi
- Molecular Pharmacology, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Via Amadeo 42, 20133 Milan, Italy.
| | - Nadia Zaffaroni
- Molecular Pharmacology, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Via Amadeo 42, 20133 Milan, Italy.
| |
Collapse
|
49
|
Shi ZM, Wang L, Shen H, Jiang CF, Ge X, Li DM, Wen YY, Sun HR, Pan MH, Li W, Shu YQ, Liu LZ, Peiper SC, He J, Jiang BH. Downregulation of miR-218 contributes to epithelial-mesenchymal transition and tumor metastasis in lung cancer by targeting Slug/ZEB2 signaling. Oncogene 2017; 36:2577-2588. [PMID: 28192397 PMCID: PMC5422710 DOI: 10.1038/onc.2016.414] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 08/03/2016] [Accepted: 09/02/2016] [Indexed: 12/15/2022]
Abstract
Epithelial–mesenchymal transition (EMT) has been recognized as a key element of cell migration and invasion in lung cancer; however, the underlying mechanisms are not fully elucidated. Recently, emerging evidence suggest that miRNAs have crucial roles in control of EMT and EMT-associated traits such as migration, invasion and chemoresistance. Here, we found that miR-218 expression levels were significantly downregulated in lung cancer tissues compared with adjacent non-cancerous tissues, and the levels of miR-218 were significantly associated with histological grades and lymph node metastasis. Overexpression of miR-218 inhibited cell migration and invasion as well as the EMT process. Of particular importance, miR-218 was involved in the metastatic process of lung cancer cells in vivo by suppressing local invasion and distant colonization. We identified Slug and ZEB2 as direct functional targets of miR-218. Inverse correlations were observed between miR-218 levels and Slug/ZEB2 levels in cancer tissue samples. In addition, overexpression of miR-218 in H1299 increased chemosensitivity of cells to cisplatin treatment through suppression of Slug and ZEB2. These findings highlight an important role of miR-218 in the regulation of EMT-related traits and metastasis of lung cancer in part by modulation of Slug/ZEB2 signaling, and provide a potential therapeutic strategy by targeting miR-218 in NSCLC.
Collapse
Affiliation(s)
- Z-M Shi
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,State Key Lab of Reproductive Medicine, Key Laboratory of Human Functional Genomics of Jiangsu Province, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention, and Treatment, Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - L Wang
- State Key Lab of Reproductive Medicine, Key Laboratory of Human Functional Genomics of Jiangsu Province, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention, and Treatment, Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - H Shen
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - C-F Jiang
- State Key Lab of Reproductive Medicine, Key Laboratory of Human Functional Genomics of Jiangsu Province, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention, and Treatment, Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - X Ge
- State Key Lab of Reproductive Medicine, Key Laboratory of Human Functional Genomics of Jiangsu Province, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention, and Treatment, Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - D-M Li
- State Key Lab of Reproductive Medicine, Key Laboratory of Human Functional Genomics of Jiangsu Province, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention, and Treatment, Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Y-Y Wen
- State Key Lab of Reproductive Medicine, Key Laboratory of Human Functional Genomics of Jiangsu Province, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention, and Treatment, Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - H-R Sun
- State Key Lab of Reproductive Medicine, Key Laboratory of Human Functional Genomics of Jiangsu Province, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention, and Treatment, Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - M-H Pan
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - W Li
- Department of Pathology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Y-Q Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - L-Z Liu
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - S C Peiper
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - J He
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - B-H Jiang
- State Key Lab of Reproductive Medicine, Key Laboratory of Human Functional Genomics of Jiangsu Province, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention, and Treatment, Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing, China.,Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
50
|
Jafri MA, Al-Qahtani MH, Shay JW. Role of miRNAs in human cancer metastasis: Implications for therapeutic intervention. Semin Cancer Biol 2017; 44:117-131. [PMID: 28188828 DOI: 10.1016/j.semcancer.2017.02.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/03/2017] [Accepted: 02/06/2017] [Indexed: 12/23/2022]
Abstract
Metastasis is the spread and growth of localized cancer to new locations in the body and is considered the main cause of cancer-related deaths. Metastatic cancer cells display distinct genomic and epigenomic profiles and almost universally an aggressive pathophysiology. A better understanding of the molecular mechanisms and regulation of metastasis, including how metastatic tumors grow and survive in the nascent niche and the interactions of the emergent metastatic cancer cells within the local microenvironment may provide tools to design strategies to restrict metastatic dissemination. Aberrant microRNAs (miRNA) expression has been reported in metastatic cancer cells. MicroRNAs are known to regulate divergent and/or convergent metastatic gene pathways including activation of reprogramming switches during metastasis. An in-depth understanding of role of miRNAs in the metastatic cascade may lead to the identification of novel targets for anti-metastatic therapeutics as well as potential candidate miRNAs for cancer treatment. This review primarily focuses on the role of miRNAs in the mechanisms of cancer metastasis as well as implications for metastatic cancer treatment.
Collapse
Affiliation(s)
- Mohammad Alam Jafri
- Center of Excellence for Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | | | - Jerry William Shay
- Center of Excellence for Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Department of Cell Biology, University of Texas, Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|