1
|
D'Aprile S, Denaro S, Lavoro A, Candido S, Giallongo S, Torrisi F, Salvatorelli L, Lazzarino G, Amorini AM, Lazzarino G, Magro G, Tibullo D, Libra M, Giallongo C, Vicario N, Parenti R. Glioblastoma mesenchymal subtype enhances antioxidant defence to reduce susceptibility to ferroptosis. Sci Rep 2024; 14:20770. [PMID: 39237744 PMCID: PMC11377710 DOI: 10.1038/s41598-024-72024-8] [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: 04/23/2024] [Accepted: 09/03/2024] [Indexed: 09/07/2024] Open
Abstract
Glioblastoma (GBM) represents an aggressive brain tumor, characterized by intra- and inter-tumoral heterogeneity and therapy resistance, leading to unfavourable prognosis. An increasing number of studies pays attention on the regulation of ferroptosis, an iron-dependent cell death, as a strategy to reverse drug resistance in cancer. However, the debate on whether this strategy may have important implications for the treatment of GBM is still ongoing. In the present study, we used ferric ammonium citrate and erastin to evaluate ferroptosis induction effects on two human GBM cell lines, U-251 MG, with proneural characteristics, and T98-G, with a mesenchymal profile. The response to ferroptosis induction was markedly different between cell lines, indeed T98-G cells showed an enhanced antioxidant defence, with increased glutathione levels, as compared to U-251 MG cells. Moreover, using bioinformatic approaches and analysing publicly available datasets from patients' biopsies, we found that GBM with a mesenchymal phenotype showed an up-regulation of several genes involved in antioxidant mechanisms as compared to proneural subtype. Thus, our results suggest that GBM subtypes differently respond to ferroptosis induction, emphasizing the significance of further molecular studies on GBM to better discriminate between various tumor subtypes and progressively move towards personalized therapy.
Collapse
Affiliation(s)
- Simona D'Aprile
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Simona Denaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Alessandro Lavoro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Sebastiano Giallongo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Filippo Torrisi
- Department of Medicine and Surgery, University of Enna "Kore", 94100, Enna, Italy
| | - Lucia Salvatorelli
- Department of Medical and Surgical Sciences and Advanced Technologies, F. Ingrassia, University of Catania, 95123, Catania, Italy
| | - Giacomo Lazzarino
- Departmental Faculty of Medicine, UniCamillus-Saint Camillus International University of Health Sciences, Via Di Sant'Alessandro 8, 00131, Rome, Italy
| | - Angela Maria Amorini
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Gaetano Magro
- Department of Medical and Surgical Sciences and Advanced Technologies, F. Ingrassia, University of Catania, 95123, Catania, Italy
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Cesarina Giallongo
- Department of Medical and Surgical Sciences and Advanced Technologies, F. Ingrassia, University of Catania, 95123, Catania, Italy.
| | - Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy.
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| |
Collapse
|
2
|
Wang S, Cheng M, Wang S, Jiang W, Yang F, Shen X, Zhang L, Yan X, Jiang B, Fan K. A Self-Catalytic NO/O 2 Gas-Releasing Nanozyme for Radiotherapy Sensitization through Vascular Normalization and Hypoxia Relief. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403921. [PMID: 39101290 DOI: 10.1002/adma.202403921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/29/2024] [Indexed: 08/06/2024]
Abstract
Radiotherapy (RT), essential for treating various cancers, faces challenges from tumor hypoxia, which induces radioresistance. A tumor-targeted "prosthetic-Arginine" coassembled nanozyme system, engineered to catalytically generate nitric oxide (NO) and oxygen (O2) in the tumor microenvironment (TME), overcoming hypoxia and enhancing radiosensitivity is presented. This system integrates the prosthetic heme of nitric oxide synthase (NOS) and catalase (CAT) with NO-donating Fmoc-protected Arginine and Ru3+ ions, creating HRRu nanozymes that merge NOS and CAT functionalities. Surface modification with human heavy chain ferritin (HFn) improves the targeting ability of nanozymes (HRRu-HFn) to tumor tissues. In the TME, strategic arginine incorporation within the nanozyme allows autonomous O2 and NO release, triggered by endogenous hydrogen peroxide, elevating NO and O2 levels to normalize vasculature and improve blood perfusion, thus mitigating hypoxia. Employing the intrinsic O2-transporting ability of heme, HRRu-HFn nanozymes also deliver O2 directly to the tumor site. Utilizing esophageal squamous cell carcinoma as a tumor model, the studies reveal that the synergistic functions of NO and O2 production, alongside targeted delivery, enable the HRRu-HFn nanozymes to combat tumor hypoxia and potentiate radiotherapy. This HRRu-HFn nanozyme based approach holds the potential to reduce the radiation dose required and minimize side effects associated with conventional radiotherapy.
Collapse
Affiliation(s)
- Shuyu Wang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Miaomiao Cheng
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Shenghui Wang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Wei Jiang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Feifei Yang
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, 330022, China
| | - Xiaomei Shen
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, 330022, China
| | - Lirong Zhang
- State Key Laboratory of Esophageal Cancer Prevention &Treatment, Henan, 450001, China
| | - Xiyun Yan
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Jiang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
| | - Kelong Fan
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
3
|
Yan Z, Duan C, Li X, Wang H, Li S, Zhou X, Miao Y. circ-TFRC downregulation suppresses ovarian cancer progression via miR-615-3p/IGF2 axis regulation. Cancer Cell Int 2024; 24:152. [PMID: 38678242 PMCID: PMC11056059 DOI: 10.1186/s12935-024-03287-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 02/29/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Ovarian cancer (OC) is a malignancy among female globally. Circular RNAs (circRNAs) are a family of circular endogenous RNAs generated from selective splicing, which take part in many traits. Former investigation suggested that circ-TFRC was abnormally expressed in breast cancer (BC). Further, the role of circ-TFRC to the progress of OC remains unclear. So, the aim of this study was to reveal the regulatory mechanism of circ-TFRC. METHODS Our team made the luciferase reporter assay to validate circ-TFRC downstream target. Transwell migration assay, 5-ethynyl-20-deoxyuridine, and cell counting kit-8 were applied to investigate both proliferation and migration. In vivo tumorigenesis and metastasis assays were performed to investigate the circ-TFRC role in OC. RESULTS The outputs elucidated that circ-TFRC expression incremented in OC cells and tissues. circ-TFRC downregulation inhibited OC cell proliferation as well as migration in in vivo and in vitro experiments. The luciferase results validated that miR-615-3p and IGF2 were circ-TFRC downstream targets. IGF2 overexpression or miR-615-3p inhibition reversed OC cell migration after circ-TFRC silencing. Also, IGF2 overexpression reversed OC cell migration and proliferation post miR-615-3p upregulation. CONCLUSION Results demonstrate that circ-TFRC downregulation inhibits OC progression and metastasis via IGF2 expression regulation and miR-615-3psponging.
Collapse
Affiliation(s)
- Zhongxin Yan
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China.
| | - Changling Duan
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Xi Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Hao Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Shanji Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Xuexin Zhou
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Yi Miao
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
4
|
D'Aprile S, Denaro S, Pavone AM, Giallongo S, Giallongo C, Distefano A, Salvatorelli L, Torrisi F, Giuffrida R, Forte S, Tibullo D, Li Volti G, Magro G, Vicario N, Parenti R. Anaplastic thyroid cancer cells reduce CD71 levels to increase iron overload tolerance. J Transl Med 2023; 21:780. [PMID: 37924062 PMCID: PMC10625232 DOI: 10.1186/s12967-023-04664-9] [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: 10/04/2023] [Accepted: 10/25/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Follicular thyroid cancer (FTC) is a prevalent form of differentiated thyroid cancer, whereas anaplastic thyroid cancer (ATC) represents a rare, fast-growing, undifferentiated, and highly aggressive tumor, posing significant challenges for eradication. Ferroptosis, an iron-dependent cell death mechanism driven by the excessive production of reactive oxygen species and subsequent lipid peroxidation, emerges as a promising therapeutic strategy for cancer. It has been observed that many cancer cells exhibit sensitivity to ferroptosis, while some other histotypes appear to be resistant, by counteracting the metabolic changes and oxidative stress induced by iron overload. METHODS Here we used human biopsies and in vitro approaches to analyse the effects of iron-dependent cell death. We assessed cell proliferation and viability through MTT turnover, clonogenic assays, and cytofluorimetric-assisted analysis. Lipid peroxidation assay and western blot were used to analyse molecular mechanisms underlying ferroptosis modulation. Two distinct thyroid cancer cell lines, FTC-133 (follicular) and 8505C (anaplastic), were utilized. These cell lines were exposed to ferroptosis inducers, Erastin and RSL3, while simulating an iron overload condition using ferric ammonium citrate. RESULTS Our evidence suggests that FTC-133 cell line, exposed to iron overload, reduced their viability and showed increased ferroptosis. In contrast, the 8505C cell line seems to better tolerate ferroptosis, responding by modulating CD71, which is involved in iron internalization and seems to have a role in resistance to iron overload and consequently in maintaining cell viability. CONCLUSIONS The differential tolerance to ferroptosis observed in our study may hold clinical implications, particularly in addressing the unmet therapeutic needs associated with ATC treatment, where resistance to ferroptosis appears more pronounced compared to FTC.
Collapse
Affiliation(s)
- Simona D'Aprile
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Simona Denaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Anna Maria Pavone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Sebastiano Giallongo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Cesarina Giallongo
- Department of Medical and Surgical Sciences and Advanced Technologies, F. Ingrassia, University of Catania, 95123, Catania, Italy
| | - Alfio Distefano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Lucia Salvatorelli
- Department of Medical and Surgical Sciences and Advanced Technologies, F. Ingrassia, University of Catania, 95123, Catania, Italy
| | - Filippo Torrisi
- Medicine and Surgery, University of Enna "Kore", 94100, Enna, Italy
| | | | | | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Gaetano Magro
- Department of Medical and Surgical Sciences and Advanced Technologies, F. Ingrassia, University of Catania, 95123, Catania, Italy
| | - Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy.
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy.
| |
Collapse
|
5
|
Feng G, Arima Y, Midorikawa K, Kobayashi H, Oikawa S, Zhao W, Zhang Z, Takeuchi K, Murata M. Knockdown of TFRC suppressed the progression of nasopharyngeal carcinoma by downregulating the PI3K/Akt/mTOR pathway. Cancer Cell Int 2023; 23:185. [PMID: 37644594 PMCID: PMC10466839 DOI: 10.1186/s12935-023-02995-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/19/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND The transferrin receptor (TfR) encoded by TFRC gene is the main cellular iron importer. TfR is highly expressed in many cancers and is expected to be a promising new target for cancer therapy; however, its role in nasopharyngeal carcinoma (NPC) remains unknown. METHODS The TfR levels were investigated in NPC tissues and cell lines using immunohistochemistry and reverse transcription-quantitative polymerase chain reaction. Knockdown of TFRC using two siRNA to investigate the effects on intracellular iron level and biological functions, including proliferation by CKK-8 assay, colony formation, cell apoptosis and cell cycle by flow cytometry, migration and invasion, and tumor growth in vivo by nude mouse xenografts. RNA sequencing was performed to find possible mechanism after TFRC knockdown on NPC cells and further verified by western blotting. RESULTS TfR was overexpressed in NPC cell lines and tissues. Knockdown of TFRC inhibited cell proliferation concomitant with increased apoptosis and cell cycle arrest, and it decreased intracellular iron, colony formation, migration, invasion, and epithelial-mesenchymal transition in HK1-EBV cells. Western blotting showed that TFRC knockdown suppressed the levels of the iron storage protein FTH1, anti-apoptotic marker BCL-xL, and epithelial-mesenchymal transition markers. We confirmed in vivo that TFRC knockdown also inhibited NPC tumor growth and decreased Ki67 expression in tumor tissues of nude mouse xenografts. RNA sequencing and western blotting revealed that TFRC silencing inhibited the PI3K/Akt/mTOR signaling pathway. CONCLUSIONS These results indicated that TfR was overexpressed in NPC, and TFRC knockdown inhibited NPC progression by suppressing the PI3K/Akt/mTOR signaling pathway. Thus, TfR may serve as a novel biomarker and therapeutic target for NPC.
Collapse
Affiliation(s)
- Guofei Feng
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, 514-8507, Mie, Japan
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, Tsu, 514-8507, Mie, Japan
| | - Yasushi Arima
- Graduate School of Health Science, Suzuka University of Medical Science, Suzuka, 510-0226, Mie, Japan
| | - Kaoru Midorikawa
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, 514-8507, Mie, Japan
| | - Hatasu Kobayashi
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, 514-8507, Mie, Japan
| | - Shinji Oikawa
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, 514-8507, Mie, Japan
| | - Weilin Zhao
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zhe Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Kazuhiko Takeuchi
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, Tsu, 514-8507, Mie, Japan.
| | - Mariko Murata
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, 514-8507, Mie, Japan.
| |
Collapse
|
6
|
Wang C, Li N, Li Y, Hou S, Zhang W, Meng Z, Wang S, Jia Q, Tan J, Wang R, Zhang R. Engineering a HEK-293T exosome-based delivery platform for efficient tumor-targeting chemotherapy/internal irradiation combination therapy. J Nanobiotechnology 2022; 20:247. [PMID: 35642064 PMCID: PMC9153154 DOI: 10.1186/s12951-022-01462-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 05/16/2022] [Indexed: 12/21/2022] Open
Abstract
Exosomes are nanoscale monolayer membrane vesicles that are actively endogenously secreted by mammalian cells. Currently, multifunctional exosomes with tumor-targeted imaging and therapeutic potential have aroused widespread interest in cancer research. Herein, we developed a multifunctional HEK-293T exosome-based targeted delivery platform by engineering HEK-293T cells to express a well-characterized exosomal membrane protein (Lamp2b) fused to the αv integrin-specific iRGD peptide and tyrosine fragments. This platform was loaded with doxorubicin (Dox) and labeled with radioiodine-131 (131I) using the chloramine-T method. iRGD exosomes showed highly efficient targeting and Dox delivery to integrin αvβ3-positive anaplastic thyroid carcinoma (ATC) cells as demonstrated by confocal imaging and flow cytometry in vitro and an excellent tumor-targeting capacity confirmed by single-photon emission computed tomography-computed tomography after labeling with 131I in vivo. In addition, intravenous injection of this vehicle delivered Dox and 131I specifically to tumor tissues, leading to significant tumor growth inhibition in an 8505C xenograft mouse model, while showing biosafety and no side effects. These as-developed multifunctional exosomes (denoted as Dox@iRGD-Exos-131I) provide novel insight into the current treatment of ATC and hold great potential for improving therapeutic efficacy against a wide range of integrin αvβ3-overexpressing tumors.
Collapse
Affiliation(s)
- Congcong Wang
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao, 266003, Shandong, China
| | - Ning Li
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yutian Li
- Department of Radiology, Qingdao Women and Children's Hospital, No. 217 Liaoyang West Road, Shibei District, Qingdao, 266000, Shandong, China
| | - Shasha Hou
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Wenxin Zhang
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Zhaowei Meng
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Shen Wang
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Qiang Jia
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Jian Tan
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Renfei Wang
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Ruiguo Zhang
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
7
|
Cui P, Dai X, Liu R, Cao H. LncRNA LINC00888 upregulation predicts a worse survival of laryngeal cancer patients and accelerates the growth and mobility of laryngeal cancer cells through regulation of miR-378g/TFRC. J Biochem Mol Toxicol 2021; 35:e22878. [PMID: 34472153 DOI: 10.1002/jbt.22878] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/29/2021] [Accepted: 07/29/2021] [Indexed: 12/15/2022]
Abstract
In this study, we aimed to detect the clinical significance and potential mechanism of LINC00888 in laryngeal cancer. Data from The Cancer Genome Atlas (TCGA) afforded the expression of LINC00888 in laryngeal cancer samples. The clinical significance of LINC00888 expression in laryngeal cancer was demonstrated by χ2 , Cox analysis, and Kaplan-Meier. The downstream targets of LINC00888 were identified based on analysis from bioinformatics and laryngeal cancer-related TCGA data sets. The microRNA-378g (miR-378g)/TFRC (transferrin receptor) axis was selected and identified by qRT-PCR, Western blot analysis, and luciferase activity assays. Cell counting kit-8, colony formation, and transwell assays were applied to detect the phenotypes of laryngeal cancer cells. We observed that LINC00888 expression was notably increased in laryngeal cancer and associated with death, recurrence, and prognosis. Depletion of LINC00888 repressed the proliferative and motile abilities of laryngeal cancer cells in vitro. LINC00888 was predicted to act as a competing endogenous RNA within the microRNA (miRNA)/messenger RNA (mRNA) axis based on analysis from bioinformatics and laryngeal cancer-related TCGA data sets. Interestingly, we discovered that LINC00888 functioned as an miRNA sponge to suppress the effect of miR-378g on laryngeal cancer cells behaviors, as well as positively regulate TFRC expression. Furthermore, the knockdown of TFRC strengthened the inhibitory effect of si-LINC00888 on laryngeal cancer cells' malignant properties. LINC00888 is an oncogenic lncRNA that promotes the growth and mobility of laryngeal cancer cells by controlling laryngeal cancer-related mRNA and tumor-suppressive miRNA. The LINC00888/miR-378g/TFRC pathway might lead to the development of laryngeal cancer cells and, therefore, might be a candidate therapeutic target for laryngeal cancer.
Collapse
Affiliation(s)
- Panpan Cui
- Department of ENT, People's Hospital of Rizhao, Shandong, China
| | - Xiuli Dai
- Department of Orthopedics, Central Hospital of Rizhao, Shandong, China
| | - Ruiyue Liu
- Department of Otolaryngology, Heze Municipal Hospital, Shandong, China
| | - Hanhai Cao
- Department of Otolaryngology-Head & Neck Surgery, The People's Hospital of Rizhao, Shandong, China
| |
Collapse
|
8
|
Candelaria PV, Leoh LS, Penichet ML, Daniels-Wells TR. Antibodies Targeting the Transferrin Receptor 1 (TfR1) as Direct Anti-cancer Agents. Front Immunol 2021; 12:607692. [PMID: 33815364 PMCID: PMC8010148 DOI: 10.3389/fimmu.2021.607692] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 02/15/2021] [Indexed: 12/15/2022] Open
Abstract
The transferrin receptor 1 (TfR1), also known as cluster of differentiation 71 (CD71), is a type II transmembrane glycoprotein that binds transferrin (Tf) and performs a critical role in cellular iron uptake through the interaction with iron-bound Tf. Iron is required for multiple cellular processes and is essential for DNA synthesis and, thus, cellular proliferation. Due to its central role in cancer cell pathology, malignant cells often overexpress TfR1 and this increased expression can be associated with poor prognosis in different types of cancer. The elevated levels of TfR1 expression on malignant cells, together with its extracellular accessibility, ability to internalize, and central role in cancer cell pathology make this receptor an attractive target for antibody-mediated therapy. The TfR1 can be targeted by antibodies for cancer therapy in two distinct ways: (1) indirectly through the use of antibodies conjugated to anti-cancer agents that are internalized by receptor-mediated endocytosis or (2) directly through the use of antibodies that disrupt the function of the receptor and/or induce Fc effector functions, such as antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP), or complement-dependent cytotoxicity (CDC). Although TfR1 has been used extensively as a target for antibody-mediated cancer therapy over the years, interest continues to increase for both targeting the receptor for delivery purposes and for its use as direct anti-cancer agents. This review focuses on the developments in the use of antibodies targeting TfR1 as direct anti-tumor agents.
Collapse
Affiliation(s)
- Pierre V. Candelaria
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Lai Sum Leoh
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Manuel L. Penichet
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, United States
- The Molecular Biology Institute, UCLA, Los Angeles, CA, United States
- UCLA AIDS Institute, UCLA, Los Angeles, CA, United States
| | - Tracy R. Daniels-Wells
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| |
Collapse
|
9
|
Amaral M, Charmier AJ, Afonso RA, Catarino J, Faísca P, Carvalho L, Ascensão L, Coelho JMP, Gaspar MM, Reis CP. Gold-Based Nanoplataform for the Treatment of Anaplastic Thyroid Carcinoma: A Step Forward. Cancers (Basel) 2021; 13:1242. [PMID: 33808984 PMCID: PMC8218498 DOI: 10.3390/cancers13061242] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/17/2022] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is a very rare subtype of thyroid carcinoma and one of the most lethal malignancies. Poor prognosis is mainly associated with its undifferentiated nature, inoperability, and failing to respond to the typically used therapies for thyroid cancer. Photothermal Therapy (PTT) entails using light to increase tissues' temperature, leading to hyperthermia-mediated cell death. Tumours are more susceptible to heat as they are unable to dissipate it. By using functionalized gold nanoparticles (AuNPs) that transform light energy into heat, it is possible to target the heat to the tumour. This study aims to formulate ATC-targeted AuNPs able to convert near-infrared light into heat, for PTT of ATC. Different AuNPs were synthetized and coated. Size, morphology, and surface plasmon resonances band were determined. The optimized coated-AuNPs were then functionalized with ligands to assess ATC's specificity. Safety, efficacy, and selectivity were assessed in vitro. The formulations were deemed safe when not irradiated (>70% cell viability) and selective for ATC. However, when irradiated, holo-transferrin-AuNPs were the most cytotoxic (22% of cell viability). The biodistribution and safety of this formulation was assessed in vivo. Overall, this novel formulation appears to be a highly promising approach to evaluate in a very near future.
Collapse
Affiliation(s)
- Mariana Amaral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (M.A.); (M.M.G.)
| | - Adília J. Charmier
- DREAMS, Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisbon, Portugal;
| | - Ricardo A. Afonso
- Ciências Funcionais e Alvos Terapêuticos, NOVA Medical School Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal;
- Departamento de Física, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - José Catarino
- Laboratório Veterinário, Faculdade de Medicina Veterinária—Universidade Lusófona de Humanidades e Tecnologias/DNAtech, 1749-024 Lisbon, Portugal; (J.C.); (P.F.)
| | - Pedro Faísca
- Laboratório Veterinário, Faculdade de Medicina Veterinária—Universidade Lusófona de Humanidades e Tecnologias/DNAtech, 1749-024 Lisbon, Portugal; (J.C.); (P.F.)
| | - Lina Carvalho
- Central Testing Laboratory, Campus de Santiago, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Lia Ascensão
- Centro de Estudos do Ambiente e do Mar (CESAM), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal;
| | - João M. P. Coelho
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal;
| | - M. Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (M.A.); (M.M.G.)
| | - Catarina Pinto Reis
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (M.A.); (M.M.G.)
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal;
| |
Collapse
|
10
|
Mao R, Shi L, Yan W, Li W, Li B, Li X. Anaplastic thyroid carcinoma combined with sclerosing mucoepidermoid carcinoma with eosinophilia: A case report. Medicine (Baltimore) 2020; 99:e22783. [PMID: 33080749 PMCID: PMC7572036 DOI: 10.1097/md.0000000000022783] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
RATIONALE Anaplastic thyroid carcinoma (ATC) is a rare highly aggressive thyroid malignancy. Thyroid sclerosing mucoepidermoid carcinoma with eosinophilia is also a rare low grade malignant thyroid neoplasm. To date, comorbidity of these 2 tumors in the thyroid gland has not been reported in the English literature. PATIENT CONCERNS Here, we present a case of a 67-year-old women with a 6-month history of mass of left neck. She complained of a painless mass in the right neck. DIAGNOSES Based on histopathological examination of H&E stained sections, immunohistochemical staining assay and molecular tests, the patient was diagnosed with ATC combined with sclerosing mucoepidermoid carcinoma with eosinophilia. INTERVENTIONS The patient underwent radical surgery for thyroid cancer. OUTCOMES No complications, local recurrence or metastases were observed during a 1 year and 3 months follow-up after surgery. LESSONS To the best of our knowledge, this is the first case report on ATC combined with sclerosing mucoepidermoid carcinoma with eosinophilia in the English literature. This condition can be easily misdiagnosed during thyroid fine needle cytology. Clinicians should perform morphological examination, immunohistochemistry and molecular tests on resected specimen to make a definitive diagnosis.
Collapse
|
11
|
Huang Y, Huang J, Huang Y, Gan L, Long L, Pu A, Xie R. TFRC promotes epithelial ovarian cancer cell proliferation and metastasis via up-regulation of AXIN2 expression. Am J Cancer Res 2020; 10:131-147. [PMID: 32064157 PMCID: PMC7017730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/06/2019] [Indexed: 06/10/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is the most common cause of gynecological cancer death. Recent studies have reported that iron overload could accelerate cancer progression. TFRC is an important participant in intracellular iron transport, and we noticed that it was abnormally overexpressed in EOC; however, its specific role in EOC remained unclear. Therefore, our study aimed to reveal the clinical significance and biological function of TFRC in human EOC. First, we detected dramatically increased TFRC expression in EOC tissues, which was associated with a worse prognosis for patients. Subsequently, we verified that TFRC knockdown significantly inhibited the proliferation and metastasis of EOC cells (SKOV3 and A2780) in vitro and in vivo. More significantly, we demonstrated that TFRC-mediated proliferation and metastasis of EOC cells resulted from its positive regulation of AXIN2 expression. In conclusion, our findings suggest that TFRC accelerates the progression of EOC by promoting cancer cell proliferation and metastasis via upregulation of AXIN2 expression, which highlights its potential as a novel therapeutic target for human EOC.
Collapse
Affiliation(s)
- Yake Huang
- Department of Obstetrics and Gynecology, Xinqiao Hospital, Army Medical UniversityChongqing 400037, China
| | - Jiani Huang
- Institute of Cancer, Xinqiao Hospital, Army Medical UniversityChongqing 400037, China
| | - Yan Huang
- Department of Obstetrics and Gynecology, Xinqiao Hospital, Army Medical UniversityChongqing 400037, China
| | - Lei Gan
- Department of Obstetrics and Gynecology, Xinqiao Hospital, Army Medical UniversityChongqing 400037, China
| | - Ling Long
- Department of Obstetrics and Gynecology, Xinqiao Hospital, Army Medical UniversityChongqing 400037, China
| | - Aimin Pu
- Department of Obstetrics and Gynecology, Xinqiao Hospital, Army Medical UniversityChongqing 400037, China
| | - Rongkai Xie
- Department of Obstetrics and Gynecology, Xinqiao Hospital, Army Medical UniversityChongqing 400037, China
| |
Collapse
|
12
|
Seyed Abutorabi E, Irani S, Yaghmaie M, Ghaffari SH. Abemaciclib (CDK4/6 Inhibitor) Blockade Induces Cytotoxicity in Human Anaplastic Thyroid Carcinoma Cells. Rep Biochem Mol Biol 2020; 8:438-445. [PMID: 32582803 PMCID: PMC7275834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 08/09/2019] [Indexed: 06/11/2023]
Abstract
BACKGROUND Thyroid cancer is the most prevalent endocrine malignancies globally. Anaplastic thyroid carcinoma (ATC) accounts for 1-3% of all Thyroid cancer. The evidence showed that ATC is a highly invasive solid tumor with poor prognosis. Despite conventional chemotherapy treatments, a considerable number of patients show developing resistance to therapeutic agents and tumor relapse. The aim of this study was the investigation anti-tumor effect of Abemaciclib (novel targeted cancer therapy drug) on Anaplastic Thyroid carcinoma SW1736 and C643 cell lines. METHODS SW1736 and C643 cell lines were treated by desire concentrations of Abemaciclib (0, 1, 2.5, 5, 10, and 20 μM) and cell viability was measured by MTT assay. Also, Anoikis resistance assay was conducted for non-adherent the cells in the exposure of Abemaciclib. The gene expression of apoptotic and anti-apoptotic genes was conducted by quantitative Real-time PCR. RESULTS Abemaciclib at the concentration of 10 and 20 μM effectively reduced cell proliferation and growth of the ATC cells compared to the control (p=0.000). Furthermore, we showed that 10 and 20 μM doses of the Abemaciclib inhibited the non-adherent ATC cells which were resistant to Anoikis death significantly (p=0.001). Moreover, we demonstrated this targeted therapy significantly reduced anti-apoptotic gene expression levels (BCL2 and CMYC) (p<0.05) and increased apoptotic gene expressions such as P21 and BAX (p<0.05). CONCLUSION Our data suggested that Abemaciclib can be utilized as a novel therapeutic agent in ATC cancer. Further in vivo and in vitro investigations are needed to evaluate molecular and clinical mechanisms of Abemaciclib.
Collapse
Affiliation(s)
- Elaheh Seyed Abutorabi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Shiva Irani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Marjan Yaghmaie
- Hematology/Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyed Hamid Ghaffari
- Hematology/Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Calabrese G, Dolcimascolo A, Caruso G, Forte S. miR-19a Is Involved In Progression And Malignancy Of Anaplastic Thyroid Cancer Cells. Onco Targets Ther 2019; 12:9571-9583. [PMID: 32009794 PMCID: PMC6859471 DOI: 10.2147/ott.s221733] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/13/2019] [Indexed: 12/15/2022] Open
Abstract
Background MicroRNAs (miRNAs) are endogenous, single-stranded, non-coding RNAs acting as negative regulators of gene expression involved in a number of physiological processes. MiRNAs' expression is commonly dysregulated in many types of human tumor diseases and cancers, including thyroid cancers, and is often involved in tumor initiation and progression. miR-19a, a member of miR-17-92 cluster, has been demonstrated to promote cell growth in anaplastic thyroid cancer (ATC), the most advanced and aggressive thyroid cancer. Purpose In this work, we investigate the potential contribution of miR-19a in thyroid cancer cells poor prognosis and de-differentiation. Methods We directly modulated the expression of miR-19a in papillary (PTC) and anaplastic thyroid carcinoma cell lines through transfection of specific miR-19a mimic or inhibitor. Further, we performed gene expression analysis of specific genes to evaluate miR-19a association with cell cycle, differentiation, and poor prognosis. Results Our data indicate that miR-19a overexpression in PTC cells significantly promotes cell growth, decreases the expression of differentiation genes and activates poor prognosis genes. Its inhibition in ATC cells reduces cell proliferation and the expression of genes related to poor prognosis but does not affect differentiation. Conclusion Our findings reveal the existence of functional associations between miR-19a expression and thyroid cancer progression and malignancy suggesting miR-19a as a novel candidate therapeutic target for ATC.
Collapse
Affiliation(s)
- Giovanna Calabrese
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania 95123, Italy
| | - Anna Dolcimascolo
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania 95123, Italy
| | - Giuseppe Caruso
- Department of Laboratories, Oasi Research Institute, IRCCs, Troina, EN 94018, Italy
| | - Stefano Forte
- Molecular Biology Unit, IOM Ricerca, Viagrande, CT 95029, Italy
| |
Collapse
|
14
|
Calabrese G, Dolcimascolo A, Torrisi F, Zappalà A, Gulino R, Parenti R. MiR-19a Overexpression in FTC-133 Cell Line Induces a More De-Differentiated and Aggressive Phenotype. Int J Mol Sci 2018; 19:ijms19123944. [PMID: 30544640 PMCID: PMC6320980 DOI: 10.3390/ijms19123944] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/29/2018] [Accepted: 12/06/2018] [Indexed: 02/07/2023] Open
Abstract
In recent years, microRNAs (miRNAs) have received increasing attention for their important role in tumor initiation and progression. MiRNAs are a class of endogenous small non-coding RNAs that negatively regulate the expression of several oncogenes or tumor suppressor genes. MiR-19a, a component of the oncogenic miR-17-92 cluster, has been reported to be highly expressed only in anaplastic thyroid cancer, the most undifferentiated, aggressive and lethal form of thyroid neoplasia. In this work, we evaluated the putative contribution of miR-19a in de-differentiation and aggressiveness of thyroid tumors. To this aim, we induced miR-19a expression in the well-differentiated follicular thyroid cancer cell line and evaluated proliferation, apoptosis and gene expression profile of cancer cells. Our results showed that miR-19a overexpression stimulates cell proliferation and alters the expression profile of genes related to thyroid cell differentiation and aggressiveness. These findings not only suggest that miR-19a has a possible involvement in de-differentiation and malignancy, but also that it could represent an important prognostic indicator and a good therapeutic target for the most aggressive thyroid cancer.
Collapse
Affiliation(s)
- Giovanna Calabrese
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania 95123, Italy.
| | - Anna Dolcimascolo
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania 95123, Italy.
| | - Filippo Torrisi
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania 95123, Italy.
| | - Agata Zappalà
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania 95123, Italy.
| | - Rosario Gulino
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania 95123, Italy.
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania 95123, Italy.
| |
Collapse
|
15
|
Ke Y, Xiang C. Transferrin receptor-targeted HMSN for sorafenib delivery in refractory differentiated thyroid cancer therapy. Int J Nanomedicine 2018; 13:8339-8354. [PMID: 30584304 PMCID: PMC6289230 DOI: 10.2147/ijn.s187240] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Thyroid cancer becomes the most common endocrine cancer with the greatest growing incidence in this decade. Sorafenib is a multikinase inhibitor for the treatment of progressive radioactive iodine-refractory differentiated thyroid cancer (DTC), while the off-target toxicity effect is usually inconvenient for patients taking. Methods In this study, hollow mesoporous silica nanoparticles (HMSNs) with transferrin modification (Tf-HMSNs) were loaded with sorafenib (sora@Tf-HMSNs) to help targeted delivery of sorafenib. Due to the biocompatible Tf shell, Tf-HMSNs exhibited excellent bio-compatibility and increased intracellular accumulation, which improved the targeting capability to cancer cells in vitro and in vivo. Results Sora@Tf-HMSNs treatment exhibited the strongest inhibition effect of res-TPC-1 cells and res-BCPAP cells compared with sora@HMSNs and sorafenib groups and induced more cancer cell apoptosis. Finally, Western blot analysis was conducted to check the expression of RAF/MEK/ERK signaling pathway after sorafenib encapsulated Tf-HMSNs treatment. Conclusion Overall, sora@Tf-HMSNs can significantly increase the effective drug concentration in cancer cells and thus enhance the anticancer effect, which are expected to be promising nanocarriers to deliver anticancer drugs for effective and safe therapy for RAI-refractory DTC.
Collapse
Affiliation(s)
- You Ke
- Department of Nephrology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Cheng Xiang
- Department of Surgery, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China,
| |
Collapse
|
16
|
Schmohl KA, Dolp P, Schug C, Knoop K, Klutz K, Schwenk N, Bartenstein P, Nelson PJ, Ogris M, Wagner E, Spitzweg C. Reintroducing the Sodium-Iodide Symporter to Anaplastic Thyroid Carcinoma. Thyroid 2017; 27:1534-1543. [PMID: 29032724 DOI: 10.1089/thy.2017.0290] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Anaplastic thyroid carcinoma (ATC), the most aggressive form of thyroid cancer, is unresponsive to radioiodine therapy. The current study aimed to extend the diagnostic and therapeutic application of radioiodine beyond the treatment of differentiated thyroid cancer by targeting the functional sodium-iodide symporter (NIS) to ATC. METHODS The study employed nanoparticle vectors (polyplexes) based on linear polyethylenimine (LPEI), shielded by polyethylene glycol (PEG) and coupled to the synthetic peptide GE11 as an epidermal growth factor receptor (EGFR)-specific ligand in order to target a NIS-expressing plasmid (LPEI-PEG-GE11/NIS) to EGFR overexpressing human thyroid carcinoma cell lines. Using ATC xenograft mouse models, transfection efficiency by 123I scintigraphy and potential for systemic radioiodine therapy after systemic polyplex application were evaluated. RESULTS In vitro iodide uptake studies in SW1736 and Hth74 ATC cells, and, for comparison, in more differentiated follicular (FTC-133) and papillary (BCPAP) thyroid carcinoma cells demonstrated high transfection efficiency and EGFR-specificity of LPEI-PEG-GE11/NIS that correlated well with EGFR expression levels. After systemic polyplex injection, in vivo 123I gamma camera imaging revealed significant tumor-specific accumulation of radioiodine in an SW1736 and an Hth74 xenograft mouse model. Radioiodine accumulation was found to be higher in SW1736 tumors, reflecting in vitro results, EGFR expression levels, and results from ex vivo analysis of NIS staining. Administration of 131I in LPEI-PEG-GE11/NIS-treated SW1736 xenograft mice resulted in significantly reduced tumor growth associated with prolonged survival compared to control animals. CONCLUSIONS The data open the exciting prospect of NIS-mediated radionuclide imaging and therapy of ATC after non-viral reintroduction of the NIS gene. The high tumor specificity after systemic application makes the strategy an attractive alternative for the treatment of highly metastatic ATC.
Collapse
Affiliation(s)
- Kathrin A Schmohl
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Patrick Dolp
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Christina Schug
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Kerstin Knoop
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Kathrin Klutz
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Nathalie Schwenk
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Peter Bartenstein
- 2 Department of Nuclear Medicine, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Peter J Nelson
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Manfred Ogris
- 3 Department of Pharmaceutical Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna , Vienna, Austria
| | - Ernst Wagner
- 4 Department of Pharmaceutical Biotechnology, Department of Pharmacy, Center for System-Based Drug Research and Center for Nanoscience , LMU Munich, Munich, Germany
| | - Christine Spitzweg
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| |
Collapse
|
17
|
Gigliotti CL, Ferrara B, Occhipinti S, Boggio E, Barrera G, Pizzimenti S, Giovarelli M, Fantozzi R, Chiocchetti A, Argenziano M, Clemente N, Trotta F, Marchiò C, Annaratone L, Boldorini R, Dianzani U, Cavalli R, Dianzani C. Enhanced cytotoxic effect of camptothecin nanosponges in anaplastic thyroid cancer cells in vitro and in vivo on orthotopic xenograft tumors. Drug Deliv 2017; 24:670-680. [PMID: 28368209 PMCID: PMC8241155 DOI: 10.1080/10717544.2017.1303856] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/02/2017] [Accepted: 03/05/2017] [Indexed: 12/30/2022] Open
Abstract
Anaplastic carcinoma of the thyroid (ATC) is a lethal human malignant cancer with median survival of 6 months. To date, no treatment has substantially changed its course, which makes urgent need for the development of novel drugs or novel formulations for drug delivery. Nanomedicine has enormous potential to improve the accuracy of cancer therapy by enhancing availability and stability, decreasing effective doses and reducing side effects of drugs. Camptothecin (CPT) is an inhibitor of DNA topoisomerase-I with several anticancer properties but has poor solubility and a high degradation rate. Previously, we reported that CPT encapsulated in β-cyclodextrin-nanosponges (CN-CPT) increased solubility, was protected from degradation and inhibited the growth of prostate tumor cells both in vitro and in vivo. The aim of this study was to extend that work by assessing the CN-CPT effectiveness on ATC both in vitro and in vivo. Results showed that CN-CPT significantly inhibited viability, clonogenic capacity and cell-cycle progression of ATC cell lines showing a faster and enhanced effect compared to free CPT. Moreover, CN-CPT inhibited tumor cell adhesion to vascular endothelial cells, migration, secretion of pro-angiogenic factors (IL-8 and VEGF-α), expression of β-PIX, belonging to the Rho family activators, and phosphorylation of the Erk1/2 MAPK. Finally, CN-CPT significantly inhibited the growth, the metastatization and the vascularization of orthotopic ATC xenografts in SCID/beige mice without apparent toxic effects in vivo. This work extends the previous insight showing that β-cyclodextrin-nanosponges are a promising tool for the treatment of ATC.
Collapse
Affiliation(s)
- Casimiro Luca Gigliotti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, UPO, Novara, Italy
| | - Benedetta Ferrara
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Sergio Occhipinti
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Elena Boggio
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, UPO, Novara, Italy
| | - Giuseppina Barrera
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Mirella Giovarelli
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Roberto Fantozzi
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, UPO, Novara, Italy
| | - Monica Argenziano
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Nausicaa Clemente
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, UPO, Novara, Italy
| | - Francesco Trotta
- Department of Chemistry, University of Torino, Torino, Italy, and
| | - Caterina Marchiò
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Laura Annaratone
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Renzo Boldorini
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, UPO, Novara, Italy
| | - Umberto Dianzani
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, UPO, Novara, Italy
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Chiara Dianzani
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| |
Collapse
|
18
|
Kirschner LS, Qamri Z, Kari S, Ashtekar A. Mouse models of thyroid cancer: A 2015 update. Mol Cell Endocrinol 2016; 421:18-27. [PMID: 26123589 PMCID: PMC4691568 DOI: 10.1016/j.mce.2015.06.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 05/28/2015] [Accepted: 06/11/2015] [Indexed: 02/06/2023]
Abstract
Thyroid cancer is the most common endocrine neoplasm, and its rate is rising at an alarming pace. Thus, there is a compelling need to develop in vivo models which will not only enable the confirmation of the oncogenic potential of driver genes, but also point the way towards the development of new therapeutics. Over the past 20 years, techniques for the generation of mouse models of human diseases have progressed substantially, accompanied by parallel advances in the genetics and genomics of human tumors. This convergence has enabled the development of mouse lines carrying mutations in the genes that cause thyroid cancers of all subtypes, including differentiated papillary and follicular thyroid cancers, poorly differentiated/anaplastic cancers, and medullary thyroid cancers. In this review, we will discuss the state of the art of mouse modeling of thyroid cancer, with the eventual goal of providing insight into tumor biology and treatment.
Collapse
Affiliation(s)
- Lawrence S Kirschner
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA; Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH, USA.
| | - Zahida Qamri
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH, USA
| | - Suresh Kari
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH, USA
| | - Amruta Ashtekar
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
19
|
Hemphill J, Borchardt EK, Brown K, Asokan A, Deiters A. Optical Control of CRISPR/Cas9 Gene Editing. J Am Chem Soc 2015; 137:5642-5. [PMID: 25905628 DOI: 10.1021/ja512664v] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The CRISPR/Cas9 system has emerged as an important tool in biomedical research for a wide range of applications, with significant potential for genome engineering and gene therapy. In order to achieve conditional control of the CRISPR/Cas9 system, a genetically encoded light-activated Cas9 was engineered through the site-specific installation of a caged lysine amino acid. Several potential lysine residues were identified as viable caging sites that can be modified to optically control Cas9 function, as demonstrated through optical activation and deactivation of both exogenous and endogenous gene function.
Collapse
Affiliation(s)
- James Hemphill
- †Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | | | - Kalyn Brown
- †Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | | | - Alexander Deiters
- †Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
20
|
Champa D, Di Cristofano A. Modeling anaplastic thyroid carcinoma in the mouse. Discov Oncol 2014; 6:37-44. [PMID: 25420535 DOI: 10.1007/s12672-014-0208-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 11/12/2014] [Indexed: 02/07/2023] Open
Abstract
Anaplastic thyroid carcinoma is the least common form of thyroid cancer; however, it accounts for the majority of deaths associated with this family of malignancies. A number of genetically engineered immunocompetent mouse models recapitulating the genetic and histological features of anaplastic thyroid cancer have been very recently generated and represent an invaluable tool to dissect the mechanisms involved in the progression from indolent, well-differentiated tumors to aggressive, undifferentiated carcinomas and to identify novel therapeutic targets. In this review, we focus on the relevant characteristics associated with these models and on what we have learned in terms of anaplastic thyroid cancer biology, genetics, and response to targeted therapy.
Collapse
Affiliation(s)
- Devora Champa
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Room 302, Bronx, NY, 10461, USA
| | | |
Collapse
|