1
|
Zhang W, Li M, Zhao Z, Xu J, Liu J, Feng P, Zhang B, Huang Z, Kong QQ, Lin Y. Tetrahedral Framework Nucleic Acid-Loaded Retinoic Acid Promotes Osteosarcoma Stem Cell Clearance. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58452-58463. [PMID: 39425646 DOI: 10.1021/acsami.4c14440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Metastatic osteosarcoma is a commonly seen malignant tumor in adolescents, with a five year survival rate of approximately 20% and a lack of treatment options. Osteosarcoma cancer stem cells are considered to be important drivers of the metastasis of osteosarcoma, and therefore their clearance is considered a promising strategy for treating metastatic osteosarcoma. In the relevant literature, retinoic acid (ATRA) is considered effective for eliminating osteosarcoma stem cells, but it has some inherent disadvantages, including poor solubility, difficulty in entering cells, and structural instability. Tetrahedral framework nucleic acids (tFNAs) are a type of nanoparticles that can carry small-molecule drugs into cells to exert therapeutic effects. Therefore, we designed and synthesized a nanoparticle named T-ATRA by using tFNAs to load ATRA and studied its effect in a nude mouse model. T-ATRA is more effective than ATRA in the clearance of osteosarcoma stem cells and in inhibiting osteosarcoma cell metastasis via the Wnt signaling pathway, thus prolonging the survival time of nude mice with osteosarcoma.
Collapse
Affiliation(s)
- Weifei Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Mengqing Li
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Zhen Zhao
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiangshan Xu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Junlin Liu
- Department of Orthopedics Surgery, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan 610041, China
| | - Pin Feng
- Department of Orthopedics Surgery, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan 610041, China
| | - Bin Zhang
- Department of Orthopedics Surgery, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan 610041, China
| | - Zhangheng Huang
- Department of Orthopaedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Qing-Quan Kong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Orthopedics Surgery, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| |
Collapse
|
2
|
Yuan B, Shi K, Zha J, Cai Y, Gu Y, Huang K, Yue W, Zhai Q, Ding N, Ren W, He W, Xu Y, Wang T. Nuclear receptor modulators inhibit osteosarcoma cell proliferation and tumour growth by regulating the mTOR signaling pathway. Cell Death Dis 2023; 14:51. [PMID: 36681687 PMCID: PMC9867777 DOI: 10.1038/s41419-022-05545-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 01/22/2023]
Abstract
Osteosarcoma is the most common primary malignant bone tumour in children and adolescents. Chemoresistance leads to poor responses to conventional therapy in patients with osteosarcoma. The discovery of novel effective therapeutic targets and drugs is still the main focus of osteosarcoma research. Nuclear receptors (NRs) have shown substantial promise as novel therapeutic targets for various cancers. In the present study, we performed a drug screen using 29 chemicals that specifically target 17 NRs in several different human osteosarcoma and osteoblast cell lines. The retinoic acid receptor beta (RARb) antagonist LE135, peroxisome proliferator activated receptor gamma (PPARg) antagonist T0070907, liver X receptor (LXR) agonist T0901317 and Rev-Erba agonist SR9011 significantly inhibited the proliferation of malignant osteosarcoma cells (U2OS, HOS-MNNG and Saos-2 cells) but did not inhibit the growth of normal osteoblasts. The effects of these NR modulators on osteosarcoma cells occurred in a dose-dependent manner and were not observed in NR-knockout osteosarcoma cells. These NR modulators also significantly inhibited osteosarcoma growth in vivo and enhanced the antitumour effect of doxorubicin (DOX). Transcriptomic and immunoblotting results showed that these NR modulators may inhibit the growth of osteosarcoma cells by regulating the PI3K/AKT/mTOR and ERK/mTOR pathways. DDIT4, which blocks mTOR activation, was identified as one of the common downstream target genes of these NRs. DDIT4 knockout significantly attenuated the inhibitory effects of these NR modulators on osteosarcoma cell growth. Together, our results revealed that modulators of RARb, PPARg, LXRs and Rev-Erba inhibit osteosarcoma growth both in vitro and in vivo through the mTOR signaling pathway, suggesting that treatment with these NR modulators is a novel potential therapeutic strategy.
Collapse
Affiliation(s)
- Baoshi Yuan
- Cambridge-Su Genomic Resource Center, Suzhou medical college of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Kexin Shi
- Cambridge-Su Genomic Resource Center, Suzhou medical college of Soochow University, Suzhou, Jiangsu, 215123, China
- Ministry of Education Key Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310030, China
| | - Juanmin Zha
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Yujia Cai
- Cambridge-Su Genomic Resource Center, Suzhou medical college of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yue Gu
- Cambridge-Su Genomic Resource Center, Suzhou medical college of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Kai Huang
- Cambridge-Su Genomic Resource Center, Suzhou medical college of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Wenchang Yue
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Qiaocheng Zhai
- Department of Orthopaedics, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China
| | - Ning Ding
- Cambridge-Su Genomic Resource Center, Suzhou medical college of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Wenyan Ren
- Cambridge-Su Genomic Resource Center, Suzhou medical college of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Weiqi He
- Cambridge-Su Genomic Resource Center, Suzhou medical college of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Ying Xu
- Cambridge-Su Genomic Resource Center, Suzhou medical college of Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Tao Wang
- Cambridge-Su Genomic Resource Center, Suzhou medical college of Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
3
|
Song J, Yuan X, Piao L, Wang J, Wang P, Zhuang M, Liu J, Liu Z. Cellular functions and molecular mechanisms of ubiquitination in osteosarcoma. Front Oncol 2022; 12:1072701. [DOI: 10.3389/fonc.2022.1072701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
Although some advances have been made in the treatment of osteosarcoma in recent years, surgical resection remains the mainstream treatment. Initial and early diagnosis of osteosarcoma could be very difficult to achieve due to the insufficient sensitivity for the means of examination. The distal metastasis of osteosarcoma also predicts the poor prognosis of osteosarcoma. In order to solve this series of problems, people begin to discover a new method of diagnosing and treating osteosarcoma. Ubiquitination, as an emerging posttranslational modification, has been shown to be closely related to osteosarcoma in studies over the past decades. In general, this review describes the cellular functions and molecular mechanisms of ubiquitination during the development of osteosarcoma.
Collapse
|
4
|
Liu H, Craig SEL, Molchanov V, Floramo JS, Zhao Y, Yang T. SUMOylation in Skeletal Development, Homeostasis, and Disease. Cells 2022; 11:cells11172710. [PMID: 36078118 PMCID: PMC9454984 DOI: 10.3390/cells11172710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/19/2022] [Accepted: 08/27/2022] [Indexed: 11/18/2022] Open
Abstract
The modification of proteins by small ubiquitin-related modifier (SUMO) molecules, SUMOylation, is a key post-translational modification involved in a variety of biological processes, such as chromosome organization, DNA replication and repair, transcription, nuclear transport, and cell signaling transduction. In recent years, emerging evidence has shown that SUMOylation regulates the development and homeostasis of the skeletal system, with its dysregulation causing skeletal diseases, suggesting that SUMOylation pathways may serve as a promising therapeutic target. In this review, we summarize the current understanding of the molecular mechanisms by which SUMOylation pathways regulate skeletal cells in physiological and disease contexts.
Collapse
Affiliation(s)
| | | | | | | | | | - Tao Yang
- Laboratory of Skeletal Biology, Department of Cell Biology, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
- Correspondence: ; Tel.: +1-616-234-5820
| |
Collapse
|
5
|
Bar-Hai N, Ishay-Ronen D. Engaging plasticity: Differentiation therapy in solid tumors. Front Pharmacol 2022; 13:944773. [PMID: 36034865 PMCID: PMC9410762 DOI: 10.3389/fphar.2022.944773] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer is a systemic heterogeneous disease that can undergo several rounds of latency and activation. Tumor progression evolves by increasing diversity, adaptation to signals from the microenvironment and escape mechanisms from therapy. These dynamic processes indicate necessity for cell plasticity. Epithelial-mesenchymal transition (EMT) plays a major role in facilitating cell plasticity in solid tumors by inducing dedifferentiation and cell type transitions. These two practices, plasticity and dedifferentiation enhance tumor heterogeneity creating a key challenge in cancer treatment. In this review we will explore cancer cell plasticity and elaborate treatment modalities that aspire to overcome such dynamic processes in solid tumors. We will further discuss the therapeutic potential of utilizing enhanced cell plasticity for differentiation therapy.
Collapse
Affiliation(s)
- Neta Bar-Hai
- Cancer Research Center, Oncology Institute, Chaim Sheba Medical Center, Tel-Hashomer, Israel
- Affiliated with Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dana Ishay-Ronen
- Cancer Research Center, Oncology Institute, Chaim Sheba Medical Center, Tel-Hashomer, Israel
- Affiliated with Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- *Correspondence: Dana Ishay-Ronen,
| |
Collapse
|
6
|
Li HB, Huang G, Tu J, Lv DM, Jin QL, Chen JK, Zou YT, Lee DF, Shen JN, Xie XB. METTL14-mediated epitranscriptome modification of MN1 mRNA promote tumorigenicity and all-trans-retinoic acid resistance in osteosarcoma. EBioMedicine 2022; 82:104142. [PMID: 35810559 PMCID: PMC9272358 DOI: 10.1016/j.ebiom.2022.104142] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 11/29/2022] Open
Abstract
Background Osteosarcoma (OS) is the most common primary malignant bone tumor in adolescents. The molecular mechanism behind OS progression and metastasis remains poorly understood, which limits the effectiveness of current therapies. RNA N6-methyladenosine (m6A) modification plays a critical role in influencing RNA fate. However, the biological significance of m6A modification and its potential regulatory mechanisms in the development of OS remain unclear. Methods Liquid chromatography-tandem mass spectrometry (LC-MS/MS), dot blotting, and colorimetric ELISA were used to detect m6A levels. Western blotting, quantitative real-time PCR (RT-qPCR) and immunohistochemistry (IHC) were used to investigate METTL14 expression levels. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and transcriptomic RNA sequencing (RNA-seq) were used to screen the target genes of METTL14. RNA pull-down and RNA immunoprecipitation (RIP) assays were conducted to explore the specific binding of target genes and relevant m6A “readers”. RNA stability and polysome analysis assays were used to detect the half-lives and translation efficiencies of the downstream genes of METTL14. IHC and clinical data were applied to explore the clinical correlations of METTL14 and its downstream target genes with the prognosis of OS. Findings We observed the abundance of m6A modifications in OS and revealed that METTL14 plays an oncogenic role in facilitating OS progression. MeRIP-seq and RNA-seq revealed that MN1 is a downstream gene of METTL14. MN1 contributes to tumor progression and all-trans-retinoic acid (ATRA) chemotherapy resistance in OS. Mechanistically, MN1 is methylated by METTL14, specifically in the coding sequence (CDS) regions, and this modification is recognized by the specific m6A reader insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) to prevent MN1 mRNA degradation and promote it translation efficiency. IHC showed that MN1 expression was positively correlated with METTL14 and IGF2BP2 expression in OS tissues. The METTL14-IGF2BP2-MN1 panel demonstrated more promising prognostic value for OS patients than any of these molecules individually. Interpretation Our study revealed that METTL14 contributes to OS progression and ATRA resistance as an m6A RNA methylase by regulating the stability and translation efficiency of MN1 and thus provides both an underlying biomarker panel for prognosis prediction in OS patients. Funding This work was supported by the National Natural Science Foundation of China (Grants 81972510 and 81772864).
Collapse
Affiliation(s)
- Hong-Bo Li
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Gang Huang
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Jian Tu
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Dong-Ming Lv
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Qing-Lin Jin
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Jun-Kai Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Yu-Tong Zou
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Dung-Fang Lee
- Department of Integrative Biology & Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, TX 77030-1501, US
| | - Jing-Nan Shen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China.
| | - Xian-Biao Xie
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
7
|
Ruh M, Stemmler MP, Frisch I, Fuchs K, van Roey R, Kleemann J, Roas M, Schuhwerk H, Eccles RL, Agaimy A, Baumhoer D, Berx G, Müller F, Brabletz T, Brabletz S. The EMT transcription factor ZEB1 blocks osteoblastic differentiation in bone development and osteosarcoma. J Pathol 2021; 254:199-211. [PMID: 33675037 DOI: 10.1002/path.5659] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 01/30/2021] [Accepted: 03/03/2021] [Indexed: 12/20/2022]
Abstract
Osteosarcoma is an often-fatal mesenchyme-derived malignancy in children and young adults. Overexpression of EMT-transcription factors (EMT-TFs) has been associated with poor clinical outcome. Here, we demonstrated that the EMT-TF ZEB1 is able to block osteoblastic differentiation in normal bone development as well as in osteosarcoma cells. Consequently, overexpression of ZEB1 in osteosarcoma characterizes poorly differentiated, highly metastatic subgroups and its depletion induces differentiation of osteosarcoma cells. Overexpression of ZEB1 in osteosarcoma is frequently associated with silencing of the imprinted DLK-DIO3 locus, which encodes for microRNAs targeting ZEB1. Epigenetic reactivation of this locus in osteosarcoma cells reduces ZEB1 expression, induces differentiation, and sensitizes to standard treatment, thus indicating therapeutic options for ZEB1-driven osteosarcomas. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Manuel Ruh
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Marc P Stemmler
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Isabell Frisch
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Kathrin Fuchs
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Ruthger van Roey
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Kleemann
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Maike Roas
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Harald Schuhwerk
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Rebecca L Eccles
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Abbas Agaimy
- Institute of Pathology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Daniel Baumhoer
- Bone Tumor Reference Centre, Institute of Pathology, University Hospital and University of Basel, Basel, Switzerland
| | - Geert Berx
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium.,Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Fabian Müller
- Department of Medicine 5 for Hematology and Oncology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN, Erlangen University Hospital, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Simone Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
8
|
Inhibition of retinoic acid receptor α phosphorylation represses the progression of triple-negative breast cancer via transactivating miR-3074-5p to target DHRS3. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:141. [PMID: 33902658 PMCID: PMC8074416 DOI: 10.1186/s13046-021-01941-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/05/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Retinoids are promising agents in the treatment of different types of neoplasia including estrogen receptor-positive breast cancers, whereas refractoriness/low sensitivity is observed in triple-negative breast cancer (TNBC) subtype. However, the reason for these diverse retinoid-sensitivity remains elusive. METHODS Determinants of retinoid sensitivity were investigated using immunohistochemistry of primary patient samples, and identified retinoic acid receptor α (RARα) as a putative factor. The anti-tumor activity of hypo-phosphorylated RARα was investigated in TNBC cell models and a xenograft mouse model. Next, miRNA sequencing analysis was performed to identify the target miRNA of RARα, and luciferase reporter was used to confirm the direct target gene of miR-3074-5p. RESULTS We discovered that serine-77 residue of RARα was constantly phosphorylated, which correlated with TNBC's resistance to retinoids. Overexpression of a phosphorylation-defective mutant RARαS77A mimicked activated RARα and repressed TNBC cell progression both in vitro and in vivo, via activating cell cycle arrest, apoptosis, and cytotoxic autophagy, independent of RARα agonists. We further revealed that the anti-tumor action of RARαS77A was, at least in part, mediated by the up-regulation of miR-3074-5p, which directly targeted DHRS3, a reductase negatively associated with TNBC patient survival. Our results suggest that the inhibition of RARαS77 phosphorylation by either expressing RARαS77A or inhibiting RARα's phosphokinase CDK7, can bypass RA stimuli to transactivate tumor-suppressive miR-3074-5p and reduce oncogenic DHRS3, thus overcoming the RA-resistance of TNBC. CONCLUSION The novel regulatory network, involving RARαS77 phosphorylation, miR-3074-5p, and DHRS3, emerges as a new target for TNBC treatment.
Collapse
|
9
|
Xu A, Zhang N, Cao J, Zhu H, Yang B, He Q, Shao X, Ying M. Post-translational modification of retinoic acid receptor alpha and its roles in tumor cell differentiation. Biochem Pharmacol 2020; 171:113696. [DOI: 10.1016/j.bcp.2019.113696] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 11/05/2019] [Indexed: 12/22/2022]
|
10
|
Chen Y, Cao J, Zhang N, Yang B, He Q, Shao X, Ying M. Advances in differentiation therapy for osteosarcoma. Drug Discov Today 2019; 25:497-504. [PMID: 31499188 DOI: 10.1016/j.drudis.2019.08.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/04/2019] [Accepted: 08/28/2019] [Indexed: 02/08/2023]
Abstract
Differentiation therapy involves the use of agents that can induce differentiation in cancer cells, with the irreversible loss of tumour phenotype. The application of differentiation therapy in osteosarcoma has made progress because of a better understanding of the potential links between differentiation defects and tumorigenesis. Here, we review recent studies on differentiation therapy for osteosarcoma, describing a variety of differentiation inducers. By highlighting these examples of drug-induced osteosarcoma cell differentiation, we can acquire unique insights into not only osteosarcoma treatment, but also novel approaches to transform differentiating drugs into more effective therapies for other solid tumours.
Collapse
Affiliation(s)
- Yingqian Chen
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ji Cao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ning Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xuejing Shao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Meidan Ying
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
11
|
A Carotenoid Extract from a Southern Italian Cultivar of Pumpkin Triggers Nonprotective Autophagy in Malignant Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7468538. [PMID: 29430284 PMCID: PMC5752993 DOI: 10.1155/2017/7468538] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 10/10/2017] [Accepted: 10/24/2017] [Indexed: 12/25/2022]
Abstract
Carotenoids, including β-carotene, lycopene, and derivatives, such as retinoic acid, have been studied for their significant antiproliferative and differentiating activity on cancer cells in experimental models and in clinics. We are presenting here data on the mechanism of action of a carotenoid-enriched extract obtained from the pumpkin Cucurbita moschata, variety “long of Naples,” on two malignant human cell lines, Caco-2 and SAOs, derived from a colon adenocarcinoma and an osteosarcoma, respectively. The carotenoid extract has been obtained from pumpkin pulp and seeds by supercritical CO2 extraction and employed to prepare oil-in-water nanoemulsions. The nanoemulsions, applied at a final carotenoid concentration of 200–400 μg/ml, were not cytotoxic, but induced a delay in cell growth of about 40% in both SAOs and Caco-2 cell lines. This effect was associated with the activation of a “nonprotective” form of autophagy and, in SAOs cells, to the induction of cell differentiation via a mechanism that involved AMPK activation. Our data suggest the presence of a pool of bioactive compounds in the carotenoid-enriched extract, acting additively, or synergistically, to delay cell growth in cancer cells.
Collapse
|
12
|
Ubiquitination of nuclear receptors. Clin Sci (Lond) 2017; 131:917-934. [PMID: 28473472 DOI: 10.1042/cs20160708] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/26/2017] [Accepted: 01/31/2017] [Indexed: 12/17/2022]
Abstract
Nuclear receptors (NRs) are cellular proteins, which upon ligand activation, act to exert regulatory control over transcription and subsequent expression. Organized via systemic classification into seven subfamilies, NRs partake in modulating a vast expanse of physiological functions essential for maintenance of life. NRs display particular characteristics towards ubiquitination, the process of addition of specific ubiquitin tags at appropriate locations. Orchestrated through groups of enzymes harboring a diverse array of specialized structural components, the ubiquitination process emphatically alters the fate or downstream effects of NRs. Such influence is especially prominent in transcriptional processes such as promoter clearing for optimization and degradation pathways eliminating or recycling targeted proteins. Ultimately, the ubiquitination of NRs carries significant implications in terms of generating pathological clinical manifestations. Increasing evidence from studies involving patients and disease models suggests a role for ubiquitinated NRs in virtually every organ system. This supports the broad repertoire of roles that NRs play in the body, including modulatory conductors, facilitators, responders to external agents, and critical constituents for pharmacological or biological interventions. This review aims to cover relevant background and mechanisms of NRs and ubiquitination, with a focus towards elucidating subsequent pathophysiology and therapeutics in clinical disorders encompassing such ubiquitinated NRs.
Collapse
|
13
|
Liu C, Ma M, Zhang J, Gui S, Zhang X, Xue S. Galangin inhibits human osteosarcoma cells growth by inducing transforming growth factor-β1-dependent osteogenic differentiation. Biomed Pharmacother 2017; 89:1415-1421. [PMID: 28340520 DOI: 10.1016/j.biopha.2017.03.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/28/2017] [Accepted: 03/09/2017] [Indexed: 11/18/2022] Open
Abstract
Osteosarcoma is the most common primary malignancy of the musculoskeletal system, and is associated with excessive proliferation and poor differentiation of osteoblasts. Currently, despite the use of traditional chemotherapy and radiotherapy, no satisfactory and effective agent has been developed to treat the disease. Herein, we found that a flavonoid natural product, galangin, could significantly attenuate human osteosarcoma cells proliferation, without causing obvious cell apoptosis. Moreover, galangin enhanced the expression of osteoblast differentiation markers (collagen type I, alkaline phosphatase, osteocalcin and osteopontin) remarkably and elevated the alkaline phosphatase activity in human osteosarcoma cells. And galangin could also attenuated osteosarcoma growth in vivo. These bioactivities of galangin resulted from its selective activation of the transforming growth factor (TGF)-β1/Smad2/3 signaling pathway, which was demonstrated by pathway blocking experiments. These findings suggested that galangin could be a promising agent to treat osteosarcoma. In addition, targeting TGF-β1 to induce osteogenic differentiation might represent a novel therapeutic strategy to treat osteosarcoma with minimal side effects.
Collapse
Affiliation(s)
- Chunhong Liu
- Department of Orthopedic Surgery, The Second People's Hospital of Wuhu, Anhui, China.
| | - Mingming Ma
- Department of Orthopedic Surgery, The People's Hospital of Fuyang, Anhui, China.
| | - Junde Zhang
- Department of Orthopedic Surgery, The Second People's Hospital of Wuhu, Anhui, China.
| | - Shaoliu Gui
- Department of Orthopedic Surgery, The Second People's Hospital of Wuhu, Anhui, China.
| | - Xiaohai Zhang
- Department of Orthopedic Surgery, The Second People's Hospital of Wuhu, Anhui, China.
| | - Shuangtao Xue
- Department of Orthopedic Surgery, The Second People's Hospital of Wuhu, Anhui, China.
| |
Collapse
|
14
|
Wu N, Li R, Meng Z, Nie M, Chen Q, He B, Deng Z, Yin L. All-trans retinoic acid restored the osteogenic ability of BMP9 in osteosarcoma through the p38 MAPK pathway. Int J Oncol 2017; 50:1363-1371. [DOI: 10.3892/ijo.2017.3910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 02/20/2017] [Indexed: 11/06/2022] Open
|
15
|
Abstract
The translationally controlled tumor protein (TCTP) is a highly conserved protein that is regulated due to a high number of extracellular stimuli. TCTP has an important role for cell cycle and normal development. On the other side, tumor reversion and malignant transformation have been associated with TCTP. TCTP has been found among the 12 genes that are differentially expressed during mouse oocyte maturation, and an overexpression of this gene was reported in a wide variety of different cancer types. Its antiapoptotic effect is indicated by the interaction with several proapoptotic proteins of the Bcl-2 family and the p53 tumor suppressor protein. In this article, we draw attention to the role of TCTP in cancer, especially, focusing on cell differentiation and tumor reversion, a biological process by which highly tumorigenic cells lose their malignant phenotype. This protein has been shown to be the most strongly downregulated protein in revertant cells compared to the parental cancer cells. Decreased expression of TCTP results either in the reprogramming of cancer cells into reversion or apoptosis. As conventional chemotherapy is frequently associated with the development of drug resistance and high toxicity, the urge for the development of new or additional scientific approaches falls into place. Differentiation therapy aims at reinducing differentiation backward to the nonmalignant cellular state. Here, different approaches have been reported such as the induction of retinoid pathways and the use of histone deacetylase inhibitors. Also, PPARγ agonists and the activation of the vitamin D receptor have been reported as potential targets in differentiation therapy. As TCTP is known as the histamine-releasing factor, antihistaminic drugs have been shown to target this protein. Antihistaminic compounds, hydroxyzine and promethazine, inhibited cell growth of cancer cells and decreased TCTP expression of breast cancer and leukemia cells. Recently, we found that two antihistaminics, levomepromazine and buclizine, inhibited cancer cell growth by direct binding to TCTP and induction of cell differentiation. These data confirmed that TCTP is an exquisite target for anticancer differentiation therapy and antihistaminics have potential to be lead compounds for the direct interaction with TCTP as new inhibitors of human TCTP and tumor growth.
Collapse
Affiliation(s)
- Ean-Jeong Seo
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Staudinger Weg 5, 55128, Mainz, Germany
| | - Nicolas Fischer
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Staudinger Weg 5, 55128, Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Staudinger Weg 5, 55128, Mainz, Germany.
| |
Collapse
|
16
|
Liu BY, Lu YQ, Han F, Wang Y, Mo XK, Han JX. Effects of the overexpression of IFITM5 and IFITM5 c.-14C>T mutation on human osteosarcoma cells. Oncol Lett 2016; 13:111-118. [PMID: 28123530 PMCID: PMC5244967 DOI: 10.3892/ol.2016.5426] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/19/2016] [Indexed: 11/18/2022] Open
Abstract
The present study aimed to investigate the effects of overexpression of interferon-induced transmembrane protein 5 (IFITM5) and IFITM5 c.-14C>T mutation on osteogenic differentiation, and the proliferation, migration and invasion of SaOS2 cells. SaOS2 cells were transfected with plasmids containing wild type IFITM5 (W) or IFITM5 containing the c.-14C>T mutation (MU). The mRNA and protein expression levels of IFITM5 in SaOS2 cells were respectively detected by reverse transcription quantitative polymerase chain reaction and western blotting. The proliferative, migratory and invasive ability of SaOS2 cells was also examined. In addition, the expression levels of osteogenic differentiation markers alkaline phosphatase (ALP), osteocalcin (OCN) and runt-related transcription factor 2 (Runx2) were detected. Mineralized nodules were detected by Alizarin Red S staining and were quantified by measuring absorbance. The mRNA and protein expression levels of IFITM5 were high in cells transfected with IFITM5 and IFITM5 c.-14C>T mutation, and were higher in cells transfected with IFITM5 c.-14C>T mutation. There was no difference in proliferation between the control group (C) and the W and MU groups. However, overexpression of IFITM5 and IFITM5 c.-14C>T mutation increased apoptotic rate, decreased invasive capacity, increased the expression of ALP, OCN and Runx2, and increased the number of mineralized nodules following osteogenic induction. In addition, compared with C and W groups, cells transfected with IFITM5 c.-14C>T mutation exhibited decreased migratory ability. In conclusion, overexpression of IFITM5 and IFITM5 c.-14C>T mutation promotes tumor cell apoptosis, inhibits tumor invasion and promotes osteogenic differentiation. These findings may provide a theoretical basis for the development of a novel treatment method that targets IFITM5, and provides a platform for the potential treatment of human osteosarcoma.
Collapse
Affiliation(s)
- Bao-Yan Liu
- Shandong Medical Biotechnological Center, School of Medicine and Life Science, Shandong Academy of Medical Sciences, University of Jinan, Jinan, Shandong 250062, P.R. China
| | - Yan-Qin Lu
- Key Laboratory for Rare Disease Research of Shandong, Shandong Medical Biotechnological Center, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Feng Han
- Shandong Medical Biotechnological Center, School of Medicine and Life Science, Shandong Academy of Medical Sciences, University of Jinan, Jinan, Shandong 250062, P.R. China
| | - Yong Wang
- Department of Neurosurgery, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xin-Kai Mo
- Shandong Medical Biotechnological Center, School of Medicine and Life Science, Shandong Academy of Medical Sciences, University of Jinan, Jinan, Shandong 250062, P.R. China; Clinical Laboratory, Shandong Cancer Hospital and Institution, Jinan, Shandong 250117, P.R. China
| | - Jin-Xiang Han
- Key Laboratory for Rare Disease Research of Shandong, Shandong Medical Biotechnological Center, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| |
Collapse
|
17
|
Seo EJ, Efferth T. Interaction of antihistaminic drugs with human translationally controlled tumor protein (TCTP) as novel approach for differentiation therapy. Oncotarget 2016; 7:16818-39. [PMID: 26921194 PMCID: PMC4941353 DOI: 10.18632/oncotarget.7605] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/19/2016] [Indexed: 01/04/2023] Open
Abstract
Translationally controlled tumor protein (TCTP) represents an exquisite target for cancer differentiation therapy, because it was most strikingly down-regulated in tumor reversion experiments. Since TCTP is identical with the histamine releasing factor, antihistamic drugs may inhibit TCTP. Indeed, antihistaminics, such as promethazine, thioridazine, perphemazine and chlorpromazine reveal antiproliferative effects. The aim of this investigation was to study antihistaminic drugs as new TCTP inhibitors to inhibit tumor growth. Levomepromazine and buclizine showed higher in silico binding affinities to TCTP among 12 different antihistaminic compounds including the control drugs, promethazine and hydroxyzine by using Autodock4 and AutodockTools-1.5.7.rc1. Recombinant human TCTP was codon-optimized, expressed in E. coli and purified by chitin affinity chromatography. For experimental validation of in silico data, we applied microscale thermophoresis. Levomepromazine bound with a Kd of 57.2 μM (p < 0.01) and buclizine with a Kd of 433μM (p < 0.01) to recombinant TCTP. Both drugs inhibited MCF-7 breast cancer cell growth in resazurin assays. TCTP expression was down-regulated after treatment with the two drugs. Cell cycle was arrested in the G1 phase without apoptosis as confirmed by the expression of cell cycle and apoptosis-regulating proteins. Annexin V-PI staining and Trypan blue exclusion assay supported that the two drugs are cytostatic rather than cytotoxic. Induction of differentiation with two drugs was detected by the increased appearance of lipid droplets. In conclusion, levomepromazine and buclizine inhibited cancer cell growth by binding to TCTP and induction of cell differentiation. These compounds may serve as lead compounds for cancer differentiation therapy.
Collapse
Affiliation(s)
- Ean-Jeong Seo
- Institute of Pharmacy and Biochemistry, Department of Pharmaceutical Biology, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Thomas Efferth
- Institute of Pharmacy and Biochemistry, Department of Pharmaceutical Biology, Johannes Gutenberg University, 55128 Mainz, Germany
| |
Collapse
|
18
|
The E3 ubiquitin protein ligase MDM2 dictates all-trans retinoic acid-induced osteoblastic differentiation of osteosarcoma cells by modulating the degradation of RARα. Oncogene 2016; 35:4358-67. [DOI: 10.1038/onc.2015.503] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 11/30/2015] [Accepted: 12/04/2015] [Indexed: 12/15/2022]
|
19
|
Sun L, Li J, Yan B. Gene expression profiling analysis of osteosarcoma cell lines. Mol Med Rep 2015; 12:4266-4272. [PMID: 26096802 PMCID: PMC4526047 DOI: 10.3892/mmr.2015.3958] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 05/13/2015] [Indexed: 12/20/2022] Open
Abstract
Osteosarcoma (OS) is the most common type of primary bone malignancy and has a poor prognosis. To investigate the mechanisms of osteosarcoma, the present analyzed the GSE28424 microarray. GSE28424 was downloaded from the Gene Expression Omnibus, and included a collective of 19 OS cell lines and four normal bone cell lines, which were used as controls. Subsequently, the differentially expressed genes (DEGs) were screened using the Limma package in Bioconductor. Gene Ontology (GO) and pathway enrichment analysis of the DEGs was performed using the Database for Annotation, Visualization and Integrated Discovery, interactions between the proteins encoded by the DEGs were identified using STRING, and the protein‑protein interaction (PPI) network was visualized using Cytoscape. In addition, modular analysis of the PPI network was performed using the Clique Percolation Method (CPM) in CFinder. A total of 1,170 DEGs were screened, including 530 upreguated and 640 downregulated genes. The enriched functions included organelle fission, immune response and response to wounding. In addition, RPL8 was observed to be involved with the ribosomal pathway in module A of the PPI network of the DEGs. PLCG1, SYK and PLCG2 were also involved in the B‑cell receptor signaling pathway in module B and the Fc‑epsilon RI signaling pathway in module C. In addition, AURKA (degree=39), MAD2L1 (degree=38), CDCA8 (degree=38), BUB1 (degree=37) and MELK (degree=37) exhibited higher degrees of connectivity in module F. The results of the present study suggested that the RPL8, PLCG1, PLCG2, SYK, MAD2L1, AURKA, CDCA8, BUB1 and MELK genes may be involved in OS.
Collapse
Affiliation(s)
- Lu Sun
- Department of Orthopedics, Shandong Chinese Medical Hospital, Jinan, Shandong 250014, P.R. China
| | - Jie Li
- Department of Orthopedics, Shandong Chinese Medical Hospital, Jinan, Shandong 250014, P.R. China
| | - Bing Yan
- Department of Orthopedics, Shandong Chinese Medical Hospital, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
20
|
Abstract
The primary bone malignancy osteosarcoma (OS) is a painful health burden, of which treatment remains a challenging problem. Identification of specific tumor biomarkers may help to investigate and develop the novel effective therapeutic approaches that have specific molecular target for the treatment of patients with OS. Osteopontin (OPN), a phosphorylated glycoprotein, is involved in many biological processes, such as biomineralization, bone remodeling and immune responses and has recently been reported to be associated with OS pathogenesis. Interestingly, both of the up- and down-regulation of OPN are involved in OS. During OS development, genetic or epigenetic disruption causes reduced expression of RUNX2 and OPN through the up-regulation of notch signaling pathway, leading to the development of OS. On the other hand, during hypoxic condition, upregulation of OPN induces the glucose uptake into hypoxic OS cells which is responsible for the OS cell proliferation and drug resistance. Recent evidences show that targeting OPN might be an important tool in OS therapeutics. This review has focused on the association of abnormal OPN expression with the pathogenesis of OS, the efficiency of OPN as a diagnostic tool for OS and the therapeutic aspects of OS by targeting OPN.
Collapse
|
21
|
Li M, Sun Y, Guan X, Shu X, Li C. Advanced progress on the relationship between RA and its receptors and malignant tumors. Crit Rev Oncol Hematol 2014; 91:271-82. [DOI: 10.1016/j.critrevonc.2014.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 03/20/2014] [Accepted: 04/02/2014] [Indexed: 12/27/2022] Open
|
22
|
Hyperoside, a flavonoid compound, inhibits proliferation and stimulates osteogenic differentiation of human osteosarcoma cells. PLoS One 2014; 9:e98973. [PMID: 24983940 PMCID: PMC4077650 DOI: 10.1371/journal.pone.0098973] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 05/09/2014] [Indexed: 11/19/2022] Open
Abstract
Osteosarcoma, one of the most common malignant bone tumours, is generally considered a differentiation disease caused by genetic and epigenetic disruptions in the terminal differentiation of osteoblasts. Novel therapies based on the non-cytotoxic induction of cell differentiation-responsive pathways could represent a significant advance in treating osteosarcoma; however, effective pharmaceuticals to induce differentiation are lacking. In the present study, we investigated the effect of hyperoside, a flavonoid compound, on the osteoblastic differentiation of U2OS and MG63 osteosarcoma cells in vitro. Our results demonstrated that hyperoside inhibits the proliferation of osteosarcoma cells by inducing G0/G1 arrest in the cell cycle, without causing obvious cell death. Cell migration assay further suggested that hyperoside could inhibit the invasion potential of osteosarcoma cells. Additionally, osteopontin and runt-related transcription factor 2 protein levels and osteocalcin activation were upregulated dramatically in hyperoside-treated osteosarcoma cells, suggesting that hyperoside may stimulates osteoblastic differentiation in osteosarcoma cells. This differentiation was accompanied by the activation of transforming growth factor (TGF)-β and bone morphogenetic protein-2, suggesting that the hyperoside-induced differentiation involves the TGF-β signalling pathway. To our knowledge, this study is the first to evaluate the differentiation effect of hyperoside in osteosarcoma cells and assess the possible potential for hyperoside treatment as a future therapeutic approach for osteosarcoma differentiation therapy.
Collapse
|
23
|
Zhou Q, Zhang L, Chen Z, Zhao P, Ma Y, Yang B, He Q, Ying M. Small ubiquitin-related modifier-1 modification regulates all-trans-retinoic acid-induced differentiation via stabilization of retinoic acid receptor α. FEBS J 2014; 281:3032-47. [DOI: 10.1111/febs.12840] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 04/17/2014] [Accepted: 05/09/2014] [Indexed: 11/30/2022]
Affiliation(s)
- Qian Zhou
- Institute of Pharmacology & Toxicology; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou China
| | - Lei Zhang
- Institute of Pharmacology & Toxicology; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou China
| | - Zibo Chen
- College of Materials Science and Engineering; Central South University of Forestry and Technology; Changsha China
| | - Pingge Zhao
- Department of Clinical Pharmacy; Yiwu Central Hospital; China
| | - Yaxi Ma
- Department of Gynecology; the Second Affiliated Hospital; School of Medicine; Zhejiang University; Hangzhou China
| | - Bo Yang
- Institute of Pharmacology & Toxicology; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou China
| | - Meidan Ying
- Institute of Pharmacology & Toxicology; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou China
| |
Collapse
|
24
|
Xie N, Zhong L, Liu L, Fang Y, Qi X, Cao J, Yang B, He Q, Ying M. Autophagy contributes to dasatinib-induced myeloid differentiation of human acute myeloid leukemia cells. Biochem Pharmacol 2014; 89:74-85. [PMID: 24607273 DOI: 10.1016/j.bcp.2014.02.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 02/20/2014] [Accepted: 02/20/2014] [Indexed: 11/17/2022]
Abstract
A breakthrough in clinical oncology was achieved as All-trans-retinoic acid (ATRA) sparked intensive differentiation therapy research. However, differentiation therapy is limited because ATRA is the sole efficient agent. Dasatinib is reported to induce myeloid differentiation of acute myeloid leukemia (AML) cells in vitro, but its mechanism remains unclear. Furthermore, the ability of dasatinib to cause differentiation of AML cells has not yet been proven. We assessed the contribution of autophagy to dasatinib-induced differentiation of AML cells. We found that dasatinib induces myeloid differentiation of AML cells accompanied with autophagy induction. Pharmacological inhibition of autophagy by 3-MA, Wortmannin, LY294002 and chloroquine block dasatinib-induced AML cell differentiation, whereas the induction of autophagy by rapamycin enhances AML cell differentiation. Our results suggest that retinoic acid receptors alpha (RARα) may not be involved in dasatinib-induced differentiation. In addition, we further illustrated that even low concentration of dasatinib can enhance ATRA-induced differentiation capability through initiation of autophagy. Taken together, we conclude that autophagy enhances the dasatinib-induced differentiation, which may provide theoretical support for developing dasatinib as a promising strategy for future differentiation therapy in AML patients.
Collapse
Affiliation(s)
- Nan Xie
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Like Zhong
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lu Liu
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yanfeng Fang
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaotian Qi
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ji Cao
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yang
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Meidan Ying
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
25
|
Geng S, Sun B, Lu R, Wang J. Coleusin factor, a novel anticancer diterpenoid, inhibits osteosarcoma growth by inducing bone morphogenetic protein-2-dependent differentiation. Mol Cancer Ther 2014; 13:1431-41. [PMID: 24723453 DOI: 10.1158/1535-7163.mct-13-0934] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Coleusin factor is a diterpenoid compound isolated from the root of a tropical plant, Coleus forskohlii. Although Coleusin factor has been reported to suppress proliferation of and induce apoptosis in several types of cancer cells, the effects of Coleusin factor on osteosarcoma and the underlying mechanism are still not fully understood. In this study, we show that Coleusin factor treatment potently inhibits the growth of osteosarcoma cells associated with G(1) cell-cycle arrest. Interestingly, apoptosis and cell death are not induced. Instead, Coleusin factor causes osteosarcoma cells to exhibit typical properties of differentiated osteoblasts, including a morphologic alteration resembling osteoblasts, the expression of osteoblast differentiation markers, elevated alkaline phosphatase activity, and increased cellular mineralization. Coleusin factor treatment significantly increases the expression of bone morphogenetic protein-2 (BMP-2), a crucial osteogenic regulator, and runt-related transcription factor 2 (RUNX2), one of the key transcription factors of the BMP pathway. When BMP-2 signaling is blocked, Coleusin factor fails to inhibit cell proliferation and to induce osteoblast differentiation. Thus, upregulation of BMP-2 autocrine is critical for Coleusin factor to induce osteoblast differentiation and exert its anticancer effects on osteosarcoma. Importantly, administration of Coleusin factor inhibits the growth of osteosarcoma xenografted in nude mice without systemic or immunologic toxicity. Osteosarcoma is a highly aggressive cancer marked by the loss of normal differentiation. Coleusin factor represents a new type of BMP-2 inducer that restores differentiation in osteosarcoma cells. It may provide a promising therapeutic strategy against osteosarcoma with minimal side effects.
Collapse
Affiliation(s)
- Shuo Geng
- Authors' Affiliations: State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Graduate University of Chinese Academy of Sciences; Department of Biology, Capital Normal University, Beijing, China; and Department of Biological Sciences, Virginia Tech, Blacksburg, VirginiaAuthors' Affiliations: State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Graduate University of Chinese Academy of Sciences; Department of Biology, Capital Normal University, Beijing, China; and Department of Biological Sciences, Virginia Tech, Blacksburg, VirginiaAuthors' Affiliations: State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Graduate University of Chinese Academy of Sciences; Department of Biology, Capital Normal University, Beijing, China; and Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia
| | - Bo Sun
- Authors' Affiliations: State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Graduate University of Chinese Academy of Sciences; Department of Biology, Capital Normal University, Beijing, China; and Department of Biological Sciences, Virginia Tech, Blacksburg, VirginiaAuthors' Affiliations: State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Graduate University of Chinese Academy of Sciences; Department of Biology, Capital Normal University, Beijing, China; and Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia
| | - Ran Lu
- Authors' Affiliations: State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Graduate University of Chinese Academy of Sciences; Department of Biology, Capital Normal University, Beijing, China; and Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia
| | - Jingze Wang
- Authors' Affiliations: State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Graduate University of Chinese Academy of Sciences; Department of Biology, Capital Normal University, Beijing, China; and Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia
| |
Collapse
|
26
|
Zhang L, Zhou Q, Zhang N, Li W, Ying M, Ding W, Yang B, He Q. E2F1 impairs all-trans retinoic acid-induced osteogenic differentiation of osteosarcoma via promoting ubiquitination-mediated degradation of RARα. Cell Cycle 2014; 13:1277-87. [PMID: 24608861 DOI: 10.4161/cc.28190] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
All-trans retinoic acid (ATRA) is a widely used differentiation drug that can effectively induce osteogenic differentiation of osteosarcoma cells, but the underlying mechanism remains elusive, which limits the clinical application for ATRA in osteosarcoma patients. In this study, we identified E2F1 as a novel regulator involved in ATRA-induced osteogenic differentiation of osteosarcoma cells. We observed that osteosarcoma cells are coupled with individual differences in the expression levels of E2F1 in patients, and E2F1 impairs ATRA-induced differentiation of osteosarcoma cells. Moreover, remarkable anti-proliferative and differentiation-inducing effects of ATRA treatment are only observed in E2F1 low to negative expressed primary osteosarcoma cultures. These results strongly suggested that E2F1 may serve as a potent indicator for the effectiveness of ATRA treatment in osteosarcoma. Interestingly, E2F1 is found to downregulate retinoic acid receptor α (RARα), a key factor determines the effectiveness of ATRA. E2F1 specifically binds to RARα and promotes its ubiquitination-mediated degradation; as a consequence, RARα-mediated differentiation is inhibited in osteosarcoma. Therefore, our studies present E2F1 as a potent biomarker, as well as a therapeutic target for ATRA-based differentiation therapeutics, and raise the hope of using differentiation-based approaches for osteosarcoma patients.
Collapse
Affiliation(s)
- Lei Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Qian Zhou
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Ning Zhang
- Department of Orthopedics; The Second Affiliated Hospital of Zhejiang University; Zhejiang University; Hangzhou, China
| | - Weixu Li
- Department of Orthopedics; The Second Affiliated Hospital of Zhejiang University; Zhejiang University; Hangzhou, China
| | - Meidan Ying
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Wanjing Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| |
Collapse
|
27
|
Sciandra M, Marino MT, Manara MC, Guerzoni C, Grano M, Oranger A, Lucarelli E, Lollini PL, Dozza B, Pratelli L, Renzo MFD, Colombo MP, Picci P, Scotlandi K. CD99 drives terminal differentiation of osteosarcoma cells by acting as a spatial regulator of ERK 1/2. J Bone Miner Res 2014; 29:1295-309. [PMID: 24677094 PMCID: PMC4255300 DOI: 10.1002/jbmr.2141] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 10/28/2013] [Accepted: 11/01/2013] [Indexed: 01/10/2023]
Abstract
Differentiation therapy is an attractive treatment for osteosarcoma (OS). CD99 is a cell surface molecule expressed in mesenchymal stem cells and osteoblasts that is maintained during osteoblast differentiation while lost in OS. Herein, we show that whenever OS cells regain CD99, they become prone to reactivate the terminal differentiation program. In differentiating conditions, CD99-transfected OS cells express osteocyte markers, halt proliferation, and largely die by apoptosis, resembling the fate of mature osteoblasts. CD99 induces ERK activation, increasing its membrane-bound/cytoplasmic form rather than affecting its nuclear localization. Through cytoplasmic ERK, CD99 promotes activity of the main osteogenic transcriptional factors AP1 and RUNX2, which in turn enhance osteocalcin and p21(WAF1/CIP1) , leading to G0 /G1 arrest. These data underscore the alternative positions of active ERK into distinct subcellular compartments as key events for determining OS fate.
Collapse
Affiliation(s)
- Marika Sciandra
- CRS Development of Biomolecular Therapies, Laboratory of Experimental Oncology, Rizzoli Orthopedic Institute, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Vourtsis D, Lamprou M, Sadikoglou E, Giannou A, Theodorakopoulou O, Sarrou E, Magoulas GE, Bariamis SE, Athanassopoulos CM, Drainas D, Papaioannou D, Papadimitriou E. Effect of an all-trans-retinoic acid conjugate with spermine on viability of human prostate cancer and endothelial cells in vitro and angiogenesis in vivo. Eur J Pharmacol 2013. [DOI: 10.1016/j.ejphar.2012.11.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
29
|
Abstract
Genetic and epigenetic events within a cell which promote a block in normal development or differentiation coupled with unregulated proliferation are hallmarks of neoplastic transformation. Differentiation therapy involves the use of agents with the ability to induce differentiation in cells that have lost this ability, i.e. cancer cells. The promise of differentiation-based therapy as a viable treatment modality is perhaps best characterized by the addition of retinoids in the treatment of acute promyelocytic leukemia (APML) revolutionizing the management of APML and dramatically improving survival. However, interest and application of differentiationbased therapy for the treatment of solid malignancies have lagged due to deficiencies in our understanding of differentiation pathways in solid malignancies. Over the past decade, a differentiation-based developmental model for solid tumors has emerged providing insights into the biology of various solid tumors as well as identification of targetable pathways capable of re-activating blocked terminal differentiation programs. Furthermore, a variety of agents including retinoids, histone deacetylase inhibitors (HDACI), PPARγ agonists, and others, currently in use for a variety of malignancies, have been shown to induce differentiation in solid tumors. Herein we discuss the relevancy of differentiation-based therapies in solid tumors, using soft tissue sarcomas (STS) as a biologic and clinical model, and review the preclinical data to support its role as a promising modality of therapy for the treatment of solid tumors.
Collapse
Affiliation(s)
- Filemon Dela Cruz
- Division of Pediatric Oncology, Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | | |
Collapse
|
30
|
Ying M, Liu G, Shimada H, Ding W, May WA, He Q, Adams GB, Wu L. Human osteosarcoma CD49f(-)CD133(+) cells: impaired in osteogenic fate while gain of tumorigenicity. Oncogene 2012; 32:4252-63. [PMID: 23045288 PMCID: PMC3947577 DOI: 10.1038/onc.2012.438] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 07/12/2012] [Accepted: 08/06/2012] [Indexed: 12/29/2022]
Abstract
The biological relationships among self-renewal, tumorigenicity, and lineage differentiation of human osteosarcoma-initiating cells (OSIC) remain elusive, making it difficult to identify and distinguish OSIC from osteosarcoma-forming cells (OSFC) for developing OSIC-targeted therapies. Using a new inverse lineage tracking strategy coupled with serial human-to-mouse xenotransplantation, we identified a subpopulation of osteosarcoma cells with OSIC-like properties and sought to distinguish them from their progeny, OSFC. We found that serial transplantation of cells from different osteosarcoma cell lines and primary osteosarcoma tissues progressively increased the CD49f+ subpopulation composing the bulk of the osteosarcoma mass. These CD49f+ cells displayed characteristics of OSFC: limited in vivo tumorigenicity, weak lineage differentiation, more differentiated osteogenic feature, and greater chemo-sensitivity. By contrast, their parental CD49f−CD133+ cells had an inhibited osteogenic fate, together with OSIC-like properties of self-renewal, strong tumorigenicity, and differentiation to CD49f+ progeny. Hence, the CD49f−CD133+ phenotype appears to identify OSIC-like cells that possess strong tumorigenicity correlated with an impaired osteogenic fate and the ability to initiate tumor growth through generation of CD49f+ progeny. These findings advance our understanding of OSIC-like properties and, for the first time, provide a much-needed distinction between OSIC and OSFC in this cancer.
Collapse
Affiliation(s)
- M Ying
- Department of Pathology, Children's Hospital Los Angeles Saban Research Institute, Los Angeles, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Chaudhry P, Yang X, Wagner M, Jong A, Wu L. Retinoid-regulated FGF8f secretion by osteoblasts bypasses retinoid stimuli to mediate granulocytic differentiation of myeloid leukemia cells. Mol Cancer Ther 2011; 11:267-76. [PMID: 22135230 DOI: 10.1158/1535-7163.mct-11-0584] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Signaling from the human hematopoietic stem cell (HSC) niche formed by osteoblastic cells regulates hematopoiesis. We previously found that retinoic acid receptor alpha (RARα), a transcription factor activated by retinoic acid (RA), mediates both granulocytic and osteoblastic differentiation. This effect depends on decreased phosphorylation of serine 77 of RARα (RARαS77) by the cyclin-dependent kinase-activating kinase (CAK) complex, a key cell-cycle regulator. In this article, we report that, by suppressing CAK phosphorylation of RARα, RA induces FGF8f to mediate osteosarcoma U2OS cell differentiation in an autocrine manner. By contrast, paracrine FGF8f secreted into osteoblast-conditioned medium by U2OS cells transduced with FGF8f or a phosphorylation-defective RARαS77 mutant, RARαS77A, bypasses RA stimuli to cross-mediate granulocytic differentiation of different types of human leukemic myeloblasts and normal primitive hematopoietic CD34(+) cells, possibly through modulating mitogen-activated protein kinase (MAPK) pathways. Further experiments using recombinant human FGF8f (rFGF8f) stimuli, antibody neutralization, and peptide blocking showed that paracrine FGF8f is required for mediating terminal leukemic myeloblast differentiation. These studies indicate a novel regulatory mechanism of granulocytic differentiation instigated by RA from the HSC niche, which links loss of CAK phosphorylation of RARα with paracrine FGF8f-mediated MAPK signaling to mediate leukemic myeloblast differentiation in the absence of RA. Therefore, these findings provide a compelling molecular rationale for further investigation of paracrine FGF8f regulation, with the intent of devising HSC niche-based FGF8f therapeutics for myeloid leukemia, with or without RA-resistance.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cell Differentiation/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cells, Cultured
- Culture Media, Conditioned/pharmacology
- Cyclin-Dependent Kinases/metabolism
- Fibroblast Growth Factor 8/genetics
- Fibroblast Growth Factor 8/metabolism
- Fibroblast Growth Factor 8/pharmacology
- Granulocytes/drug effects
- Granulocytes/metabolism
- Granulocytes/pathology
- HL-60 Cells
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Humans
- Leukemia, Myeloid/genetics
- Leukemia, Myeloid/metabolism
- Leukemia, Myeloid/pathology
- Mice
- Mice, Inbred NOD
- Mice, Nude
- Mice, SCID
- Mitogen-Activated Protein Kinases/metabolism
- Mutation
- Osteoblasts/drug effects
- Osteoblasts/metabolism
- Phosphorylation/drug effects
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/metabolism
- Retinoic Acid Receptor alpha
- Retinoids/pharmacology
- Stem Cell Niche/drug effects
- Cyclin-Dependent Kinase-Activating Kinase
Collapse
Affiliation(s)
- Parvesh Chaudhry
- Department of Pathology, Children's Hospital Los Angeles Saban Research Institute, Los Angeles, CA, USA
| | | | | | | | | |
Collapse
|