1
|
Xu T, Xu S, Ma G, Chang J, Zhang C, Zhou P, Wang C, Xu P, Yang J, Hu Y, Wu Y. Human Chorionic Gonadotropin Regulates the Smad Signaling Pathway by Antagonizing TGF-β in Giant Cell Tumor of Bone. Recent Pat Anticancer Drug Discov 2024; 19:188-198. [PMID: 38214358 PMCID: PMC10804236 DOI: 10.2174/1574892818666230413082909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND Giant cell tumor of bone (GCTB) is a locally aggressive bone tumour aggravated by stromal cell proliferation and metastasis. OBJECTIVE We investigated the mechanism of action of human chorionic gonadotropin (HCG) in mediating GCTB proliferation and invasion. METHODS The expression of HCG was quantified using quantitative real-time PCR. After the primary stromal cells were isolated and identified, the function of HCG in GCTB was estimated using the cell counting kit-8, flow cytometry, scratch experiment, transwell assay, Western blot, and immunofluorescence. Moreover, the mechanism of HCG was assessed through western blotting. RESULTS HCG expression was decreased in clinical tissue samples from patients with GCTB. We validated that HCG repressed stromal cell proliferation, migration, invasion, autophagy, and epithelial- mesenchymal transition (EMT) and promoted cell apoptosis in GCTB. We also verified that HCG repressed the autophagy and EMT of stromal cells through the Smad signaling axis in GCTB. HCG inhibited the transduction of the Smad signaling pathway by restraining the binding of the TGF-β II receptor to ligand Activin A. CONCLUSION HCG restrained the Smad signaling pathway by antagonizing TGF-β signaling in GCTB. HCG may serve as a useful patent to treat GCTB.
Collapse
Affiliation(s)
- Tangbing Xu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui Province, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, 230000, Anhui Province, China
| | - Shenglin Xu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui Province, China
| | - Guangwen Ma
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui Province, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, 230000, Anhui Province, China
| | - Jun Chang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui Province, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, 230000, Anhui Province, China
| | - Chi Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui Province, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, 230000, Anhui Province, China
| | - Ping Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui Province, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, 230000, Anhui Province, China
| | - Chao Wang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui Province, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, 230000, Anhui Province, China
| | - Pengfei Xu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui Province, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, 230000, Anhui Province, China
| | - Junjun Yang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui Province, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, 230000, Anhui Province, China
| | - Yong Hu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui Province, China
| | - Yunfeng Wu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui Province, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, 230000, Anhui Province, China
| |
Collapse
|
2
|
Wang QQ, Liu ZX, Wang P, Liu BY, Feng YP, Zhang Y, He HB, Yin T, Tang X, Wang YJ, Gou JX. Intratumoral injection of norcantharidin liposome emulsion hybrid delivery system amplifies the cancer-fighting effects of oral sorafenib against hepatocellular carcinoma. Colloids Surf B Biointerfaces 2023; 232:113599. [PMID: 37857183 DOI: 10.1016/j.colsurfb.2023.113599] [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: 08/22/2023] [Accepted: 10/14/2023] [Indexed: 10/21/2023]
Abstract
Interventional therapies are increasingly used in clinical trials for hepatocellular carcinoma (HCC). Sorafenib is the front-line remedy for HCC, however, chemoresistance occurs immutably and affects the effectiveness of treatment. In a previous study, a norcantharidin liposome emulsion hybrid (NLEH) delivery system for HCC was developed. This study aims to examine the therapeutic effects of the combination of intratumoral injection of NLEH and sorafenib in treating HCC. Sorafenib combined with NLEH activated the apoptosis pathway by synergistically upregulating caspase-9, promoting cytotoxicity, apoptosis (64.57%), and G2/M cell cycle arrest (48.96%). Norcantharidin could alleviate sorafenib resistance by counteracting sorafenib-induced phosphorylation of Akt. Additionally, intratumoral injection of NLEH exhibited a sustained accumulation in the tumor within 24 h and didn't distribute to other major organs. Intratumoral injection of NLEH in combination with oral sorafenib displayed the most potent tumor growth inhibitory effect (77.91%) in vivo. H&E staining results and the indicators of the renal and liver function tests demonstrated the safety of this combination therapy. Overall, these results showed that intratumoral injection of NLEH in combination with oral sorafenib treatment represented a rational potential therapeutic option for HCC.
Collapse
Affiliation(s)
- Qing-Qing Wang
- School of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Zi-Xu Liu
- School of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Ping Wang
- Department of Pharmaceutics, College of Pharmacy Sciences, Jilin University, Changchun, Jilin, China
| | - Bo-Yuan Liu
- School of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Yu-Peng Feng
- School of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Yu Zhang
- School of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Hai-Bing He
- School of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Tian Yin
- Department of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Xing Tang
- School of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, Liaoning, China.
| | - Yan-Jiao Wang
- School of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, Liaoning, China.
| | - Jing-Xin Gou
- School of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, Liaoning, China.
| |
Collapse
|
3
|
Tan X, Zhang Y, Wei D, Yang Y, Xiang F. Denosumab for giant cell tumors of bone from 2010 to 2022: a bibliometric analysis. Clin Exp Med 2023; 23:3053-3075. [PMID: 37103655 DOI: 10.1007/s10238-023-01079-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 04/17/2023] [Indexed: 04/28/2023]
Abstract
Giant cell tumors of the bone (GCTB) are considered moderately malignant bone tumors. Denosumab, as a neoadjuvant therapy, provides new possibilities for treating GCTB. However, even after multiple studies and long-term clinical trials, there are limitations in the treatment process. Research data and Medical Subject Headings terms related to denosumab and GCTB were collected from January 2010 to October 2022 using the Web of Science and MeSH ( https://meshb.nlm.nih.gov ) browsers. These data were imported into CiteSpace and VOSviewer softwares for bibliometric analysis. Overall, 445 publications on denosumab and GCTB were identified. Over the last 12 years, the growth rate of the total number of publications has remained relatively stable. The USA published the highest number of articles (83) and had the highest centrality (0.42). Amgen Inc. and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) First Ortoped Rizzoli were identified as the most influential institutions. Many authors have made outstanding contributions to this field. Lancet Oncology had the highest journal impact factor (54.433). Local recurrence and drug dosage are current research hotspots, and future development trends will mainly focus on prognostic markers of GCTB and the development of new therapies. Further research is required to analyze denosumab's safety and efficacy and understand its local recurrence of GCTB, to identify the optimal dose. Future progress in this field will likely focus on exploring new diagnostic and recurrence markers to monitor disease progression and examine new therapeutic targets and treatment strategies.
Collapse
Affiliation(s)
- Xiaoqi Tan
- Department of Dermatology, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yue Zhang
- Department of Orthopedic, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, China
| | - Daiqing Wei
- Department of Orthopedic, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, China
| | - Yunkang Yang
- Department of Orthopedic, Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, China.
| | - Feifan Xiang
- Department of Orthopedic, Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, China.
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China.
- Institute of Nuclear Medicine, Southwest Medical University, Luzhou, China.
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
4
|
Yousef EH, El-Magd NFA, El Gayar AM. Norcantharidin potentiates sorafenib antitumor activity in hepatocellular carcinoma rat model through inhibiting IL-6/STAT3 pathway. Transl Res 2023; 260:69-82. [PMID: 37257560 DOI: 10.1016/j.trsl.2023.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
In hepatocellular carcinoma (HCC), sorafenib (Sora) efficacy is limited by primary and/or acquired resistance. Emerging evidence shows that the inflammatory factor interleukin 6 (IL-6) plays a role in Sora resistance. Norcantharidin (NCTD), a derivative of cantharidine, was identified as a potent IL-6 inhibitor. Thus, in this study, we evaluated NCTD ability to improve the Sora efficacy in HCC and its underlying molecular mechanisms. Male Sprague Dawely rats were administered NCTD (0.1 mg/kg/day; orally) or Sora (10 mg/kg day; orally) or combination for 6 weeks after HCC induction using thioacetamide (200 mg/kg; ip; 2 times/wk) for 16 weeks. Our results showed that NCTD greatly enhanced Sora activity against HCC and potentiated Sora-induced oxidative stress. NCTD enhanced Sora-induced tumor immunity reactivation by decreasing both fibrinogen-like protein 1 level and increasing both tumor necrosis factor-α gene expression along with CD8+ T cells number. Also, NCTD augmented Sora attenuation activity against TAA-induced angiogenesis and metastasis by decreasing VEGFA, HIF-1α, serum lactate dehydrogenase enzyme, and vimentin levels. The combined use of NCTD/Sora suppressed drug resistance and stemness by downregulating ATP-binding cassette subfamily G member 2, neurogenic locus notch homolog protein, spalt-like transcription factor 4, and CD133. NCTD boosted Sora antiproliferative and apoptotic activities by decreasing Ccnd1 and BCL2 expressions along with increasing BAX and caspase-3 expressions. To our knowledge, this study represents the first study providing evidence for the potential novel therapeutic use of NCTD/Sora combination for HCC. Moreover, no previous studies have reported the effect of NCTD on FGL1.
Collapse
Affiliation(s)
- Eman H Yousef
- Biochemistry department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt; Biochemistry department, Faculty of Pharmacy, Horus University-Egypt, Damietta, Egypt.
| | - Nada F Abo El-Magd
- Biochemistry department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Amal M El Gayar
- Biochemistry department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
5
|
Hassan M, Shahzadi S, Malik A, Din SU, Yasir M, Chun W, Kloczkowski A. Oncomeric Profiles of microRNAs as New Therapeutic Targets for Treatment of Ewing's Sarcoma: A Composite Review. Genes (Basel) 2023; 14:1849. [PMID: 37895198 PMCID: PMC10606885 DOI: 10.3390/genes14101849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/07/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Ewing's sarcoma is a rare type of cancer that forms in bones and soft tissues in the body, affecting mostly children and young adults. Current treatments for ES are limited to chemotherapy and/or radiation, followed by surgery. Recently, microRNAs have shown favourable results as latent diagnostic and prognostic biomarkers in various cancers. Furthermore, microRNAs have shown to be a good therapeutic agent due to their involvement in the dysregulation of various molecular pathways linked to tumour progression, invasion, angiogenesis, and metastasis. In this review, comprehensive data mining was employed to explore various microRNAs that might have therapeutic potential as target molecules in the treatment of ES.
Collapse
Affiliation(s)
- Mubashir Hassan
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children Hospital, Columbus, OH 43205, USA;
| | - Saba Shahzadi
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children Hospital, Columbus, OH 43205, USA;
| | - Amal Malik
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 54590, Pakistan;
| | - Salah ud Din
- Department of Bioinformatics, University of Okara, Okara 56130, Pakistan;
| | - Muhammad Yasir
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea; (M.Y.); (W.C.)
| | - Wanjoo Chun
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea; (M.Y.); (W.C.)
| | - Andrzej Kloczkowski
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children Hospital, Columbus, OH 43205, USA;
- Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
| |
Collapse
|
6
|
Conrad O, Burgy M, Foppolo S, Jehl A, Thiéry A, Guihard S, Vauchelles R, Jung AC, Mourtada J, Macabre C, Ledrappier S, Chenard MP, Onea MA, Danic A, Dourlhes T, Thibault C, Schultz P, Dontenwill M, Martin S. Tumor-Suppressive and Immunomodulating Activity of miR-30a-3p and miR-30e-3p in HNSCC Cells and Tumoroids. Int J Mol Sci 2023; 24:11178. [PMID: 37446353 DOI: 10.3390/ijms241311178] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are heterogeneous tumors, well known for their frequent relapsing nature. To counter recurrence, biomarkers for early diagnosis, prognosis, or treatment response prediction are urgently needed. miRNAs can profoundly impact normal physiology and enhance oncogenesis. Among all of the miRNAs, the miR-30 family is frequently downregulated in HNSCC. Here, we determined how levels of the 3p passenger strands of miR-30a and miR-30e affect tumor behavior and clarified their functional role in LA-HNSCC. In a retrospective study, levels of miR-30a-3p and miR-30e-3p were determined in 110 patients and correlated to overall survival, locoregional relapse, and distant metastasis. miR-30a/e-3p were expressed in HNSCC cell lines and HNSCC patient-derived tumoroids (PDTs) to investigate their effect on tumor cells and their microenvironment. Both miRNAs were found to have a prognosis value since low miR-30a/e-3p expression correlates to adverse prognosis and reduces overall survival. Low expression of miR-30a/e-3p is associated with a shorter time until locoregional relapse and a shorter time until metastasis, respectively. miR-30a/e-3p expression downregulates both TGF-βR1 and BMPR2 and attenuates the survival and motility of HNSCC. Results were confirmed in PDTs. Finally, secretomes of miR-30a/e-3p-transfected HNSCC activate M1-type macrophages, which exert stronger phagocytic activities toward tumor cells. miR-30a/e-3p expression can discriminate subgroups of LA-HNSCC patients with different prognosis, making them good candidates as prognostic biomarkers. Furthermore, by targeting members of the TGF-β family and generating an immune-permissive microenvironment, they may emerge as an alternative to anti-TGF-β drugs to use in combination with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Ombline Conrad
- Laboratory of Bioimaging and Pathology, University of Strasbourg, UMR7021 CNRS, 67401 Illkirch, France
| | - Mickaël Burgy
- Laboratory of Bioimaging and Pathology, University of Strasbourg, UMR7021 CNRS, 67401 Illkirch, France
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Sophie Foppolo
- Laboratory of Bioimaging and Pathology, University of Strasbourg, UMR7021 CNRS, 67401 Illkirch, France
| | - Aude Jehl
- Laboratory of Bioimaging and Pathology, University of Strasbourg, UMR7021 CNRS, 67401 Illkirch, France
| | - Alicia Thiéry
- Department of Public Health, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Sébastien Guihard
- Department of Radiotherapy, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Romain Vauchelles
- Laboratory of Bioimaging and Pathology, University of Strasbourg, UMR7021 CNRS, 67401 Illkirch, France
| | - Alain C Jung
- Laboratory STREINTH, Inserm IRFAC U1113, Université de Strasbourg, 67200 Strasbourg, France
- Laboratory of Tumor Biology, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Jana Mourtada
- Laboratory STREINTH, Inserm IRFAC U1113, Université de Strasbourg, 67200 Strasbourg, France
| | - Christine Macabre
- Laboratory STREINTH, Inserm IRFAC U1113, Université de Strasbourg, 67200 Strasbourg, France
- Laboratory of Tumor Biology, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Sonia Ledrappier
- Laboratory STREINTH, Inserm IRFAC U1113, Université de Strasbourg, 67200 Strasbourg, France
- Laboratory of Tumor Biology, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Marie-Pierre Chenard
- Department of Pathology, Strasbourg University Hospital, 67200 Strasbourg, France
| | - Mihaela-Alina Onea
- Department of Pathology, Strasbourg University Hospital, 67200 Strasbourg, France
| | - Aurélien Danic
- Department of Otolaryngology and Cervico-Facial Surgery, Strasbourg University Hospital, 67200 Strasbourg, France
| | - Thomas Dourlhes
- Department of Otolaryngology and Cervico-Facial Surgery, Strasbourg University Hospital, 67200 Strasbourg, France
| | - Claire Thibault
- Department of Otolaryngology and Cervico-Facial Surgery, Strasbourg University Hospital, 67200 Strasbourg, France
| | - Philippe Schultz
- Department of Otolaryngology and Cervico-Facial Surgery, Strasbourg University Hospital, 67200 Strasbourg, France
| | - Monique Dontenwill
- Laboratory of Bioimaging and Pathology, University of Strasbourg, UMR7021 CNRS, 67401 Illkirch, France
| | - Sophie Martin
- Laboratory of Bioimaging and Pathology, University of Strasbourg, UMR7021 CNRS, 67401 Illkirch, France
| |
Collapse
|
7
|
Chuang YT, Tang JY, Shiau JP, Yen CY, Chang FR, Yang KH, Hou MF, Farooqi AA, Chang HW. Modulating Effects of Cancer-Derived Exosomal miRNAs and Exosomal Processing by Natural Products. Cancers (Basel) 2023; 15:318. [PMID: 36612314 PMCID: PMC9818271 DOI: 10.3390/cancers15010318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Cancer-derived exosomes exhibit sophisticated functions, such as proliferation, apoptosis, migration, resistance, and tumor microenvironment changes. Several clinical drugs modulate these exosome functions, but the impacts of natural products are not well understood. Exosome functions are regulated by exosome processing, such as secretion and assembly. The modulation of these exosome-processing genes can exert the anticancer and precancer effects of cancer-derived exosomes. This review focuses on the cancer-derived exosomal miRNAs that regulate exosome processing, acting on the natural-product-modulating cell functions of cancer cells. However, the role of exosomal processing has been overlooked in several studies of exosomal miRNAs and natural products. In this study, utilizing the bioinformatics database (miRDB), the exosome-processing genes of natural-product-modulated exosomal miRNAs were predicted. Consequently, several natural drugs that modulate exosome processing and exosomal miRNAs and regulate cancer cell functions are described here. This review sheds light on and improves our understanding of the modulating effects of exosomal miRNAs and their potential exosomal processing targets on anticancer treatments based on the use of natural products.
Collapse
Affiliation(s)
- Ya-Ting Chuang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaoshiung Medical University, Kaohsiung 80708, Taiwan
| | - Jun-Ping Shiau
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Kun-Han Yang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 54000, Pakistan
| | - Hsueh-Wei Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
8
|
Zheng J, Wang JJ, Ma HM, Shen MQ, Qian ZM, Bao YX. Norcantharidin down-regulates iron contents in the liver and spleen of lipopolysaccharide-treated mice. Redox Rep 2022; 27:119-127. [PMID: 35735222 PMCID: PMC9246006 DOI: 10.1080/13510002.2022.2088011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective The inhibiting effect of Norcantharidin (NCTD) on IL-6 (interleukin-6) and STAT3 and the involvement of the IL-6/STAT3 pathway in hepcidin expression prompted us to speculate that NCTD could affect iron metabolism. Methods We examined the effects of NCTD on serum iron (SI) and transferrin (Tf) saturation, iron and ferritin light chain (FTL), transferrin receptor 1 (TfR1), divalent metal transporter 1 (DMT1), ferroportin 1 (Fpn1), iron regulatory protein 1 (IRP1) and hepcidin, as well as IL-6 and STAT3 in the liver, spleen and duodenum of mice treated with lipopolysaccharide (LPS) in vivo, using RT-PCR, Western blotting and immunofluorescence analysis. Results NCTD could increase SI and Tf saturation and reduce tissue iron and FTL content by affecting expression of cell-iron transport proteins TfR1, DMT1 and Fpn1. The impact of NCTD on TfR1, DMT1 and Fpn1 expression is mediated by up-regulating IRP1 and down-regulating hepcidin expression, while NCTD-induced down-regulation of hepcidin is mediated by the IL-6/STAT3 signalling pathway in LPS-treated mice. Conclusions NCTD affects iron metabolism by modifying the expression of IL-6/JAK2/STAT3/hepcidin and IRP1 and suggest that the ability of NCTD to reduce tissue iron contents may be a novel mechanism associated with the anti-cancer effects of NCTD.
Collapse
Affiliation(s)
- Jie Zheng
- Research Center for Medicine and Biology, Zunyi Medical University, Zunyi, People's Republic of China
| | - Jiao-Jiao Wang
- Research Center for Medicine and Biology, Zunyi Medical University, Zunyi, People's Republic of China
| | - Hui-Min Ma
- Institute of Translational and Precision Medicine, Nantong University, Nantong, People's Republic of China
| | - Meng-Qi Shen
- Institute of Translational and Precision Medicine, Nantong University, Nantong, People's Republic of China
| | - Zhong-Ming Qian
- Institute of Translational and Precision Medicine, Nantong University, Nantong, People's Republic of China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.,Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, People's Republic of China
| | - Yu-Xin Bao
- Research Center for Medicine and Biology, Zunyi Medical University, Zunyi, People's Republic of China
| |
Collapse
|
9
|
Norcantharidin liposome emulsion hybrid delivery system enhances PD-1/PD-L1 immunotherapy by agonizing the non-canonical NF-κB pathway. Int J Pharm 2022; 628:122361. [DOI: 10.1016/j.ijpharm.2022.122361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/17/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
|
10
|
Yue J, Sun W, Li S. Denosumab Versus Zoledronic Acid in Cases of Surgically Unsalvageable Giant Cell Tumor of Bone: A Randomized Clinical Trial. J Bone Oncol 2022; 35:100441. [PMID: 35800292 PMCID: PMC9254448 DOI: 10.1016/j.jbo.2022.100441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/20/2022] [Accepted: 06/20/2022] [Indexed: 11/24/2022] Open
Abstract
Denosumab and zoledronic acid had similar tumor responses and clinical benefits. Disease progression was observed in patients in the DB group (12.5%) than ZA group (15.0%). Denosumab treatment was markedly more expensive than zoledronic acid treatment (p < 0.0001). The recurrence-free survival rate at 4-year follow-up is higher in DB group (p = 0.035).
Background Giant-cell tumor of bone (GCTB) is a relatively benign, but locally aggressive osteoclastogenic stromal tumour of the bone. Although denosumab has been approved as an monoclonal antibody against RANK ligand for the treatment of GCTB, few clinical trials of the benefit in tumor response have been conducted to prove the efficiency in Chinese population. Objectives In this multicentric, random controlled, clinical trial, 160 patients were enrolled to compare the therapeutic efficacy and safety of denosumab and zoledronic acid treatment in patients with surgically unsalvageable GCTB. Methods Between 2nd Jan 2015 and 1st Jan 2018, 160 adults (aged ≥ 18 years) with ①surgically unsalvageable GCTB, ②surgically salvageable GCTB with planned surgery expected to result in severe morbidity were included in this randomized clinical trial. Patients received either subcutaneous denosumab (DB group; 120 mg once every 4 weeks with loading doses of 120 mg subcutaneously admininstered on days 8 and 15; n = 80) or intravenous zoledronic acid (ZA group; 4 mg once every 4 weeks; n = 80) for six cycles. Disease status, clinical benefits, treatment-emergent adverse effects, overall survival, and cost of treatment were evaluated during the follow-up period. Statistical significance was determined using 95% confidence intervals. Results Denosumab and zoledronic acid had similar tumor responses (p = 0.118) and clinical benefits (p = 0.574). Disease progression was observed in fewer patients in the DB group (12.5%) than ZA group (15.0%). Denosumab caused fatigue (p = 0.001) and back pain (p < 0.0001), while zoledronic acid caused hypocalcemia (p < 0.0001), flu-like symptoms (p = 0.059) and hypotension (p = 0.059). Denosumab treatment was markedly more expensive than zoledronic acid treatment (p < 0.0001). The cost to manage treatment-emergent adverse effects was the same for the ZA group and the DB group (p = 0.425). The accumulate recurrence-free survival rate at 4-year follow-up is higher in DB group (p = 0.035). Conclusions Denosumab is a safe but costly alternative to zoledronic acid for treatment of surgically unsalvageable GCTB.
Collapse
|
11
|
Du LJ, Feng YX, He ZX, Huang L, Wang Q, Wen CP, Zhang Y. Norcantharidin ameliorates the development of murine lupus via inhibiting the generation of IL-17 producing cells. Acta Pharmacol Sin 2022; 43:1521-1533. [PMID: 34552214 PMCID: PMC9159996 DOI: 10.1038/s41401-021-00773-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a devastating autoimmune disorder associated with severe organ damage. The abnormality of T cell apoptosis is considered as an important pathogenetic mechanism of SLE. Norcantharidin (NCTD), a derivative of Cantharidin, is an efficacious anti-cancer drug by inhibiting cell proliferation and inducing cell apoptosis. Besides, NCTD has also been proved to protect the function of kidneys, while damaged renal function is the most important predictor of morbidity and mortality in SLE. All these suggest the potential effects of NCTD in SLE treatment. In this study we investigated whether NCTD exerted therapeutic effects in a mouse SLE model. Lupus prone female MRL/lpr mice were treated with NCTD (1, 2 mg·kg-1·d-1, ip) for 8 weeks. We showed that NCTD administration significantly decreased mortality rate, diminished the expression of anti-dsDNA IgG antibody, a diagnostic marker for SLE, as well as restored renal structure and function in MRL/lpr mice. Moreover, NCTD administration dose-dependently inhibited lymphoproliferation and T cell accumulation in the spleens of MRL/lpr mice. We further revealed that NCTD specifically inhibited DN T cell proliferation and Th17 cell differentiation both via blocking activation of signal transducer and activator of transcription 3 (STAT3) signaling pathway. On the other hand, NCTD did not affect T cell apoptosis in MRL/lpr mice. Taken together, our data suggest that NCTD may be as a promising therapeutic drug through targeting T cells for the treatment of SLE.
Collapse
Affiliation(s)
- Li-jun Du
- grid.268505.c0000 0000 8744 8924Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053 China
| | - Yu-xiang Feng
- grid.268505.c0000 0000 8744 8924Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053 China
| | - Zhi-xing He
- grid.268505.c0000 0000 8744 8924Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053 China
| | - Lin Huang
- grid.268505.c0000 0000 8744 8924Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053 China
| | - Qiao Wang
- grid.268505.c0000 0000 8744 8924Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053 China
| | - Cheng-ping Wen
- grid.268505.c0000 0000 8744 8924Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053 China
| | - Yun Zhang
- grid.268505.c0000 0000 8744 8924Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053 China
| |
Collapse
|
12
|
Liu Z, Zhao L, Tan X, Wu Z, Zhou N, Dong N, Zhang Y, Yin T, He H, Gou J, Tang X, Gao S. Preclinical evaluations of Norcantharidin liposome and emulsion hybrid delivery system with improved encapsulation efficiency and enhanced antitumor activity. Expert Opin Drug Deliv 2022; 19:451-464. [PMID: 35385376 DOI: 10.1080/17425247.2022.2063834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Norcantharidin (NCTD) has a certain degree of hydrophilicity and poor lipophilicity, and has some side-effects, including short t1/2, vascular irritation, cardiotoxicity and nephrotoxicity, which bring difficulties for formulation research. In this study, we aim to develop a novel nanocarrier to improve encapsulation efficiency, increase sterilization stability and enhance antitumor activity. METHODS Phospholipid complexes methods were used for increasing the lipophilicity of norcantharidin (NCTD), then NCTD phospholipid complexes were not only loaded in the oil phase and oil-water interface surface, but also encapsulated in phospholipid bilayers to obtain NCTD liposome-emulsion hybrid (NLEH) delivery system. The in vitro cytotoxicity and apoptosis, in vivo tissue distribution, tumor penetration, heterotopic and orthotopic antitumor studies were conducted to evaluate therapeutic effect. RESULTS NLEH exhibited an improved encapsulation efficiency (89.3%) and a better sterilization stability, compared to NCTD liposomes and NCTD emulsions. NLEH can achieve a better antitumor activity by promoting absorption (1.93-fold), prolonging blood circulation (2.08-fold), enhancing tumor-targeting accumulation (1.19 times), improving tumor penetration, and increasing antitumor immunity. CONCLUSIONS The liposome-emulsion hybrid (LEH) delivery system was potential carrier for NCTD delivery, and LEH could open opportunities for delivery of poorly soluble anticancer drugs, especially drugs that are more hydrophilicity than lipophilicity.
Collapse
Affiliation(s)
- Zixu Liu
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Linxuan Zhao
- Department of Pharmaceutics, College of Pharmacy Sciences, Jilin University, Changchun 130021, China
| | - Xinyi Tan
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Zixuan Wu
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Ning Zhou
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Nan Dong
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Yu Zhang
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Tian Yin
- Department of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Haibing He
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Jingxin Gou
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Xing Tang
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Song Gao
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
13
|
Xu R, Choong PFM. Metastatic giant cell tumour of bone: a narrative review of management options and approaches. ANZ J Surg 2022; 92:691-696. [PMID: 35143093 PMCID: PMC9303226 DOI: 10.1111/ans.17520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/04/2022] [Accepted: 01/17/2022] [Indexed: 11/28/2022]
Abstract
Giant cell tumour of bone (GCTB) is a locally aggressive bone neoplasm with a rare tendency to metastasise, most commonly to the lungs. The management of metastatic GCTB (metGCTB) is controversial due to its unpredictable behaviour. Asymptomatic patients should be monitored radiologically and undergo treatment only when disease progression occurs. Surgery is recommended for resectable metGCTB. Denosumab, a monoclonal antibody which inhibits receptor activator of nuclear factor-κB ligand, is recommended for unresectable metGCTB with evidence from phase II trials demonstrating its safety and efficacy. Relapse after denosumab withdrawal may occur and prolonged treatment may be associated with serious adverse events, thus further research is warranted to inform a maintenance regimen with reduced dosing and frequency. Combined denosumab and bisphosphonate therapy has the potential to achieve sustained disease control or remission in unresectable metGCTB without requiring long-term treatment and should be evaluated in prospective trials. Various novel agents have demonstrated in vitro and anecdotal efficacy and warrant further evaluation.
Collapse
Affiliation(s)
- Ruiwen Xu
- St Vincent's Hospital Clinical SchoolThe University of MelbourneMelbourneVictoriaAustralia
- Melbourne Medical SchoolThe University of MelbourneMelbourneVictoriaAustralia
| | - Peter F. M. Choong
- Department of SurgeryThe University of MelbourneMelbourneVictoriaAustralia
- Department of OrthopaedicsSt. Vincent's Hospital MelbourneMelbourneVictoriaAustralia
- Bone and Soft Tissue Sarcoma UnitPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| |
Collapse
|
14
|
Fang J, Zhu H, Xu P, Jiang R. Zingerone suppresses proliferation, invasion, and migration of hepatocellular carcinoma cells by the inhibition of MTDH-mediated PI3K/Akt pathway. J Recept Signal Transduct Res 2021; 42:409-417. [PMID: 34645355 DOI: 10.1080/10799893.2021.1988970] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE Previous studies have proved that zingerone was a therapeutic agent for many tumors. Metadherin (MTDH) acts as an oncogene and is involved in tumorigenesis. The purpose of this study was to explore the underlying mechanism of zingerone that regulates MTDH to affect hepatocellular carcinoma (HCC) progression. METHODS CCK-8 assay was performed to detect HCC cell proliferation. The invasion and migration abilities of HCC cells were evaluated using Transwell assay. The mRNA and protein levels in cells and tissues were measured using qRT-PCR and Western blot assays. Moreover, we established the HCC xenografts nude mice to evaluate the effect of zingerone on tumor growth. RESULTS We found that zingerone treatment significantly inhibited HCC cell malignant phenotype and tumor growth. Moreover, MTDH was highly expressed in HCC tissues and cell lines and was positively associated with poor overall survival of patients with HCC. Knockdown of MTDH notably suppressed the proliferation, invasion, and migration capacities of HCC cells. Mechanistically, inhibition of MTDH by zingerone impeded the malignant biological behavior of HCC cells by inactivating the PI3K/Akt pathway. CONCLUSION These results suggested that zingerone served as an effective therapeutic agent in HCC via blocking the MTDH-mediated PI3K/Akt pathway.
Collapse
Affiliation(s)
- Jian Fang
- Department of Hepatobiliary Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Huifen Zhu
- Department of Hepatobiliary Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Pengcheng Xu
- Department of Hepatobiliary Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Renya Jiang
- Department of Hepatobiliary Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| |
Collapse
|
15
|
Palmini G, Brandi ML. microRNAs and bone tumours: Role of tiny molecules in the development and progression of chondrosarcoma, of giant cell tumour of bone and of Ewing's sarcoma. Bone 2021; 149:115968. [PMID: 33892177 DOI: 10.1016/j.bone.2021.115968] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 03/26/2021] [Accepted: 04/15/2021] [Indexed: 12/15/2022]
Abstract
The increasing interest on microRNAs (miRNAs), small non-coding RNA molecules containing about 22 nucleotides, about their biological functions led researchers to discover that they are actively involved in several biological processes. In the last decades, miRNAs become one of the most topic of cancer research. miRNAs, thanks to their function, are the perfect molecules to modulate multiple signaling pathways and gene expression in cancer, with the consequent capacity to modulate cancerous processes, such as cellular proliferation, invasion, metastasis and chemoresistance in various tumours. In the last years, several studies have demonstrated the role of miRNAs in their pathophysiology, but little we know about the underlying mechanism that lead to bone tumours like chondrosarcoma (COS), giant cell tumour of bone (GCTB) and Ewing sarcoma (EWS) to still be highly aggressive and resistant tumours. An exploration of the role of miRNAs in the biology of them will permit to researchers to find new molecular mechanisms that can be used to develop new and more effective therapies against these bone tumours. Here we present a comprehensive study of the latest discoveries which have been performed in relation to the role of miRNAs in the neoplastic processes which characterize COS, EWS and GCTB, demonstrating how these tiny molecules can act as tumour promoters or as tumour suppressors and how they can be used for improving therapeutic approaches.
Collapse
Affiliation(s)
- Gaia Palmini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy.
| | - Maria Luisa Brandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy; Fondazione Italiana Ricerca sulle Malattie dell'Osso, F.I.R.M.O Onlus, Florence, Italy.
| |
Collapse
|
16
|
Lin B, Zhu J, Yin G, Liao M, Lin G, Yan Y, Huang D, Lu S. Transcription Factor DLX5 Promotes Hair Follicle Stem Cell Differentiation by Regulating the c-MYC/microRNA-29c-3p/NSD1 Axis. Front Cell Dev Biol 2021; 9:554831. [PMID: 34336814 PMCID: PMC8319474 DOI: 10.3389/fcell.2021.554831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 03/10/2021] [Indexed: 11/22/2022] Open
Abstract
Introduction Adult stem cell function has been one of the most intensively explored areas of biological and biomedical research, with hair follicle stem cells serving as one of the best model systems. This study explored the role of the transcription factor DLX5 in regulating hair follicle stem cell (HFSC) differentiation. Methods HFSCs were isolated, characterized, and assessed for their expression of DLX5, c-MYC, NSD1, and miR-29c-3p using RT-qPCR, Western blot analysis, or immunofluorescence. Next, the ability of HFSCs to proliferate as well as differentiate into either sebaceous gland cells or epidermal cells was determined. The binding of DLX5 to the c-MYC promoter region, the binding of c-MYC to the miR-29c-3p promoter region, and the binding of miR-29c-3p to the 3′-UTR of NSD1 mRNA were verified by luciferase activity assay and ChIP experiments. Results DLX5 was highly expressed in differentiated HFSCs. DLX5 transcriptionally activated c-MYC expression to induce HFSC differentiation. c-MYC was able to bind the miR-29c-3p promoter and thus suppressed its expression. Without miR-29c-3p mediated suppression, NSD1 was then able to promote HFSC differentiation. These in vitro experiments suggested that DLX5 could promote HFSC differentiation via the regulation of the c-MYC/miR-29c-3p/NSD1 axis. Discussion This study demonstrates that DLX5 promotes HFSC differentiation by modulating the c-MYC/miR-29c-3p/NSD1 axis and identifies a new mechanism regulating HFSC differentiation.
Collapse
Affiliation(s)
- Bojie Lin
- Department of Plastic and Aesthetic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiangying Zhu
- Department of Plastic and Aesthetic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Academy of Humanities and Social Sciences, Guangxi Medical University, Nanning, China
| | - Guoqian Yin
- Department of Plastic and Aesthetic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Mingde Liao
- Department of Plastic and Aesthetic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Guanyu Lin
- Department of Plastic and Aesthetic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yuyong Yan
- Department of Plastic and Aesthetic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Dan Huang
- Department of Plastic and Aesthetic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Siding Lu
- Department of Plastic and Aesthetic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
17
|
Norcantharidin protects against renal interstitial fibrosis by suppressing TWEAK-mediated Smad3 phosphorylation. Life Sci 2020; 260:118488. [PMID: 32979359 DOI: 10.1016/j.lfs.2020.118488] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 08/17/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023]
Abstract
AIMS This study investigated the role and mechanism of action of tumor necrosis factor-like weak inducer of apoptosis (TWEAK) in the pathogenesis of renal interstitial fibrosis (RIF), and its involvement in the anti-RIF effect of norcantharidin (NCTD). MAIN METHODS Mice with unilateral ureteral obstruction and BUMPT mouse proximal tubular cells exposed to transforming growth factor (TGF)-β1 were used as in vivo and in vitro models of RIF, respectively. NCTD was administered to mice by intraperitoneal injection (0.075 mg kg-1·day-1). Hematoxylin-eosin and Masson's trichrome staining were performed to assess pathologic changes in the kidney. Immunohistochemistry, western blotting, and real-time PCR were performed to evaluate the expression of TWEAK and the fibrotic factors fibronectin (FN) and collagen type I (Col-I). The role of TWEAK in RIF and in the anti-RIF effect of NCTD was evaluated by TWEAK overexpression and neutralization with a specific antibody, and specific inhibitor of Mothers against decapentaplegic homolog (Smad)3 (SIS3) was used to examine the involvement of TGF-β1/Smad3 signaling. KEY FINDINGS TWEAK was mainly expressed in renal tubules in mice; the level was markedly elevated in both in vivo and in vitro RIF models. TWEAK overexpression in BUMPT cells increased the levels of phosphorylated Smad3, FN, and Col-I, which were reduced by treatment with SIS3. NCTD suppressed FN and Col-I expression by blocking TWEAK-mediated Smad3 phosphorylation. SIGNIFICANCE Upregulation of TWEAK contributes to RIF by promoting Smad3 phosphorylation, while NCTD inhibits this process.
Collapse
|
18
|
Norcantharidin: research advances in pharmaceutical activities and derivatives in recent years. Biomed Pharmacother 2020; 131:110755. [PMID: 33152920 DOI: 10.1016/j.biopha.2020.110755] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/20/2022] Open
Abstract
Cantharidin (CTD) is the main bioactive component of Cantharides, which is called Banmao in Traditional Chinese Medicine (TCM). Norcantharidin (NCTD) is a structural modifier of CTD. To compare with CTD, NCTD has lighter side effects and stronger bioactivity in anti-cancer through inhibiting cell proliferation, causing apoptosis and autophagy, overwhelming migration and metastasis, affecting immunity as well as lymphangiogenesis. Examples of these effects include suppressing Protein Phosphatase 2A and modulating Wnt/beta catenin signal, with Caspase family proteins, AMPK pathway and c-Met/EGFR pathway involving respectively. Moreover, NCTD has the effects of immune enhancement, anti-platelet aggregation and inhibition of renal interstitial fibrosis with distinct signaling pathways. The immunological effects induced by NCTD are related to the regulation of macrophage polarization and LPS-mediated immune response. The antiplatelet activity that NCTD induced is relevant to the inhibition of platelet signaling and the downregulation of α2 integrin. Furthermore, some of novel derivatives designed and synthesized artificially show stronger biological activities (e.g., anticancer effect, enzyme inhibition effect, antioxidant effect) and lower toxicity than NCTD itself. Plenty of literatures have reported various pharmacological effects of NCTD, particularly the anticancer effect, which has been widely concerned in clinical application and laboratory research. In this review, the pharmaceutical activities and derivatives of NCTD are discussed, which can be reference for further study.
Collapse
|
19
|
Pan MS, Cao J, Fan YZ. Insight into norcantharidin, a small-molecule synthetic compound with potential multi-target anticancer activities. Chin Med 2020; 15:55. [PMID: 32514288 PMCID: PMC7260769 DOI: 10.1186/s13020-020-00338-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/25/2020] [Indexed: 02/07/2023] Open
Abstract
Norcantharidin (NCTD) is a demethylated derivative of cantharidin, which is an anticancer active ingredient of traditional Chinese medicine, and is currently used clinically as a routine anti-cancer drug in China. Clarifying the anticancer effect and molecular mechanism of NCTD is critical for its clinical application. Here, we summarized the physiological, chemical, pharmacokinetic characteristics and clinical applications of NCTD. Besides, we mainly focus on its potential multi-target anticancer activities and underlying mechanisms, and discuss the problems existing in clinical application and scientific research of NCTD, so as to provide a potential anticancer therapeutic agent for human malignant tumors.
Collapse
Affiliation(s)
- Mu-Su Pan
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Jin Cao
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Yue-Zu Fan
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| |
Collapse
|
20
|
Protocol for DNA Microarrays on Glass Slides. Methods Mol Biol 2020; 1986:17-33. [PMID: 31115883 DOI: 10.1007/978-1-4939-9442-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The DNA microarray is a powerful, flexible, nonbiased discovery technology. Microarrays can be used to assess processes from gene expression to long noncoding RNAs to specific pathologies, as well as many others. This chapter describes the protocol for DNA microarray analysis of differential gene expression using DNA sequences spotted on microscope slides.
Collapse
|
21
|
Li S, Chen P, Yang Q. Denosumab versus zoledronic acid in cases of surgically unsalvageable giant cell tumor of bone: A randomized clinical trial. J Bone Oncol 2019; 15:100217. [PMID: 30740297 PMCID: PMC6357891 DOI: 10.1016/j.jbo.2019.100217] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/16/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Although denosumab has been approved as an antiresorptive agent for giant cell tumor of bone, its efficacy has not been proven. OBJECTIVES To compare the efficacy and safety of denosumab and zoledronic acid treatment in patients with surgically unsalvageable giant cell tumor of bone. METHODS A total of 250 patients with surgically unsalvageable giant cell tumor of bone were included in this randomized clinical trial. Patients received either subcutaneous denosumab (DB group; 120 mg per 4 weeks plus an additional 120 mg on days 8 and 15; n = 125) or intravenous zoledronic acid (ZA group; 4 mg per 4 weeks; n = 125) for six cycles. Disease status, clinical benefits, treatment-emergent adverse effects, overall survival, and cost of treatment were evaluated during the follow-up period. Statistical significance was determined using 95% confidence intervals. RESULTS Denosumab and zoledronic acid had similar tumor responses (p = 0.18) and clinical benefits (p = 0.476). Disease progression was observed in fewer patients in the DB group (1%) than ZA group (2%). Denosumab caused fatigue (p = 0.0004) and back pain (p < 0.0001), while zoledronic acid caused hypocalcemia (p < 0.0001), flu-like symptoms (p = 0.021), hypotension (p = 0.021), and hypokalemia (p = 0.021). Denosumab treatment was markedly more expensive than zoledronic acid treatment (p < 0.0001). The cost to manage treatment-emergent adverse effects was higher for the ZA group than the DB group (p = 0.0425). Overall survival was the same for both treatments (p = 0.066). CONCLUSIONS Denosumab is a safe but costly alternative to zoledronic acid for treatment of surgically unsalvageable giant cell tumor of bone.
Collapse
Key Words
- ALT test, Alanine aminotransferase test
- ANOVA, analysis of variance
- AST test, Aspartate aminotransferase test
- Antiresorptive
- Bone density conservation agents
- CONSORT, Consolidated standards of reporting trials
- CT, the computed tomography
- CTCAE, Common Terminology Criteria for Adverse Events
- Denosumab
- EMA, the European Medicines Agency
- EORTC, European organization for research and treatment of cancer
- GCTB, giant cell tumor of bone
- Giant cell tumor of bone
- MRI, magnetic resonance imaging
- N, sample population
- RANKL, Receptor activator of nuclear factor kappa-Β ligand
- RECIST, response evaluation criteria in solid tumors
- US FDA, the United States Food and Drug Administration
- Zoledronic acid
- n, sample size
Collapse
Affiliation(s)
- Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No 44 of Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province 110042, China
| | | | | |
Collapse
|
22
|
He Q, Xue S, Tan Y, Zhang L, Shao Q, Xing L, Li Y, Xiang T, Luo X, Ren G. Dual inhibition of Akt and ERK signaling induces cell senescence in triple-negative breast cancer. Cancer Lett 2019; 448:94-104. [PMID: 30742942 DOI: 10.1016/j.canlet.2019.02.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/05/2019] [Accepted: 02/01/2019] [Indexed: 12/31/2022]
Abstract
Activated Akt and ERK signaling pathways are closely related to breast cancer progression, and Akt or ERK inhibition induces cell senescence. However, the crosstalk between the Akt and ERK signaling pathways in cell senescence and how to simultaneously suppress Akt and ERK signaling in triple-negative breast cancer (TNBC) are undefined. In this study, we found that norcantharidin (NCTD) effectively induced cell senescence and cell cycle arrest in TNBC in vitro, which was accompanied by a decline in phosphorylated Akt and ERK1/2 and a rise in p21 and p16. The inhibitors LY294002 and U0126 imitated the effect of NCTD when these two inhibitors were combined regardless of crosstalk between these two signaling pathways. In addition, NCTD inhibited the growth of xenografts via downregulation of phosphorylated Akt and ERK1/2 and upregulation of p21 in vivo. However, NCTD upregulated the level of soluble signaling factors of the senescence-associated secretory phenotype (SASP) in a NF-κB-independent manner. Collectively, these findings demonstrate that NCTD induced cell senescence and cell cycle arrest mainly by simultaneously blocking Akt and ERK signaling in TNBC, suggesting that NCTD may be used as a potential adjuvant therapy in TNBC.
Collapse
Affiliation(s)
- Qiang He
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | - Yiqing Tan
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Zhang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Shao
- Breast Center, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Lei Xing
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunhai Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tingxiu Xiang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinrong Luo
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|