1
|
Kögel D, Temme A, Aigner A. Recent advances in development and delivery of non-viral nucleic acid therapeutics for brain tumor therapy. Pharmacol Ther 2025; 266:108762. [PMID: 39603349 DOI: 10.1016/j.pharmthera.2024.108762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024]
Abstract
High grade gliomas (HGG) are a group of CNS tumors refractory to currently existing therapies, which routinely leads to early recurrence and a dismal prognosis. Recent advancements in nucleic acid-based therapy using a wide variety of different molecular targets and non-viral nanocarrier systems suggest that this approach holds significant potential to meet the urgent demand for improved therapeutic options for the treatment of these tumors. This review provides a comprehensive and up-to-date overview on the current landscape and progress of preclinical and clinical developments in this rapidly evolving and exciting field of research, including optimized nanocarrier delivery systems, promising therapeutic targets and tailor-made therapeutic strategies for individualized HGG patient treatment.
Collapse
Affiliation(s)
- Donat Kögel
- Department of Neurosurgery, Experimental Neurosurgery, University Hospital, Goethe University, Frankfurt am Main, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt am Main, Germany; German Cancer Research Center DKFZ, Heidelberg, Germany.
| | - Achim Temme
- Department of Neurosurgery, Section Experimental Neurosurgery/Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, Germany; National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Leipzig, Germany; Comprehensive Cancer Center Central Germany (CCCG), Site Leipzig, Leipzig, Germany
| |
Collapse
|
2
|
Wang Y, Wu H, Chen Z, Cao J, Zhu X, Zhang X. Nano-hydroxyapatite promotes cell apoptosis by co-activating endoplasmic reticulum stress and mitochondria damage to inhibit glioma growth. Regen Biomater 2024; 11:rbae038. [PMID: 38799701 PMCID: PMC11127112 DOI: 10.1093/rb/rbae038] [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: 12/30/2023] [Revised: 02/27/2024] [Accepted: 03/20/2024] [Indexed: 05/29/2024] Open
Abstract
Despite a growing body of studies demonstrating the specific anti-tumor effect of nano-hydroxyapatite (n-HA), the underlying mechanism remained unclear. Endoplasmic reticulum (ER) and mitochondria are two key players in intracellular Ca2+ homeostasis and both require Ca2+ to participate. Moreover, the ER-mitochondria interplay coordinates the maintenance of cellular Ca2+ homeostasis to prevent any negative consequences from excess of Ca2+, hence there needs in-depth study of n-HA effect on them. In this study, we fabricated needle-like n-HA to investigate the anti-tumor effectiveness as well as the underlying mechanisms from cellular and molecular perspectives. Data from in vitro experiments indicated that the growth and invasion of glioma cells were obviously reduced with the aid of n-HA. It is interesting to note that the expression of ER stress biomarkers (GRP78, p-IRE1, p-PERK, PERK, and ATF6) were all upregulated after n-HA treatment, along with the activation of the pro-apoptotic transcription factor CHOP, showing that ER stress produced by n-HA triggered cell apoptosis. Moreover, the increased expression level of intracellular reactive oxygen species and the mitochondrial membrane depolarization, as well as the downstream cell apoptotic signaling activation, further demonstrated the pro-apoptotic roles of n-HA induced Ca2+ overload through inducing mitochondria damage. The in vivo data provided additional evidence that n-HA caused ER stress and mitochondria damage in cells and effectively restrain the growth of glioma tumors. Collectively, the work showed that n-HA co-activated intracellular ER stress and mitochondria damage are critical triggers for cancer cells apoptosis, offering fresh perspectives on ER-mitochondria targeted anti-tumor therapy.
Collapse
Affiliation(s)
- Yifu Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Hongfeng Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- Medical School, Kunming University of Science and Technology, Kunming 650500, P. R. China
| | - Zhu Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- Institute of tissue engineering and stem cells, Nanchong Central Hospital, North Sichuan Medical College, Nanchong 637000, P. R. China
| | - Jun Cao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| |
Collapse
|
3
|
Nucleic acid therapy in pediatric cancer. Pharmacol Res 2022; 184:106441. [PMID: 36096420 DOI: 10.1016/j.phrs.2022.106441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/02/2022] [Accepted: 09/07/2022] [Indexed: 12/24/2022]
Abstract
The overall survival, progress free survival, and life quality of cancer patients have improved due to the advance in minimally invasive surgery, precision radiotherapy, and various combined chemotherapy in the last decade. Furthermore, the discovery of new types of therapeutics, such as immune checkpoint inhibitors and immune cell therapies have facilitated both patients and doctors to fight with cancers. Moreover, in the context of the development in biocompatible and cell type targeting nano-carriers as well as nucleic acid-based drugs for initiating and enhancing the anti-tumor response have come to the age. The treatment paradigms utilization of nucleic acids, including short interfering RNA (siRNA), antisense oligonucleotides (ASO), and messenger RNA (mRNA), can target specific protein expression to achieve the therapeutic effects. Over ten nucleic acid therapeutics have been approved by the FDA and EMA in rare diseases and genetic diseases as well as dozens of registered clinical trails for varies cancers. Though generally less dangerous of pediatric cancers than adult cancers was observed during the past decades, yet pediatric cancers accounted for a significant proportion of child deaths which hurt those family very deeply. Therefore, it is necessary to pay more attention for improving the treatment of pediatric cancer and discovering new nucleic acid therapeutics which may help to improve the therapeutic effect and prognoses in turns to ameliorate the survival period and quality of life for children patient. In this review, we focus on the nucleic acid therapy in pediatric cancers.
Collapse
|
4
|
Elfeky SA, Elsayed A, Moawad M, Ahmed WA. Hydroxyapatite nanocomposite as a potential agent in osteosarcoma PDT. Photodiagnosis Photodyn Ther 2020; 32:102056. [PMID: 33068821 DOI: 10.1016/j.pdpdt.2020.102056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/26/2020] [Accepted: 10/02/2020] [Indexed: 01/09/2023]
Abstract
Using Nanoplatforms as a hauler for photosensitizers is a bespoke paradigm to improve its bioavailability and to boost the PDT efficacy. Herein, the photodynamic cytotoxicity of methylene blue (MB) loaded on hydroxyapatite nanoparticles (HA-NPs) was tested against human osteosarcoma-derived cells (Saos-2 cell line). HA-NPs and HA-NPs loaded with MB (HA-NPs-MB) were prepared by a chemical precipitation method and characterized by TEM, Zeta potential, FTIR, and XRD. TEM images revealed that HA-NPs have a rod shape with a diameter of 14-17 nm and length around 46-64 nm. FTIR and Zeta potential confirmed the adsorption of cationic MB on HA-NPs. XRD pattern was identical to the standard XRD pattern of HA-NPs. Incubation of Saos-2 cells (24 h) with HA-NPs-MB then irradiation of cells (5 min) with a diode laser (808 nm), causes a higher decrement of cell viability (determined by MTT assay) than that caused by free MB. The LC50 was 57.53 µg/mL and 86.99 µg/mL for HA-NPs-MB and free MB, respectively. Thus, the nanoformulation of MB greatly reduced the dose of MB required for effective PDT. This study also investigated the mode of cell death after incubation of cells with free MB or HA-NPs-MB composite then exposure to laser radiation. The results revealed that the majority of cells died by apoptosis while a minor fraction of cells died by necrosis, especially in the case of HA-NPs-MB. Levels of caspase-3 and death receptor-4 (DR-4) were more elevated in the case of HA-NPs-MB than free MB. The effect of the prepared nanocomposite and free MB on Raw murine macrophage (RAW 264.7) viability was also examined using the MTT assay. The results indicated that HA-NPs-MB in the presence of laser has a great cytotoxic effect on macrophage cells compared to other treatments. This may present an advantage through decreasing macrophage that promotes tumor growth. In conclusion, HA-NPs-MB nanocomposite surmounts free MB and HA-NPs in destroying macrophage cells and Saos-2 cells through apoptosis in the presence of laser irradiation. This study introduces a thorough and new insight on osteosarcoma (cancer cell line Saos-2) PDT using HA-NPs-MB exploiting the biosafety of HA-NPs.
Collapse
Affiliation(s)
- Souad A Elfeky
- National Institute of Laser Enhanced Sciences, Cairo University, Giza, 12613, Egypt.
| | - Ahmed Elsayed
- National Institute of Laser Enhanced Sciences, Cairo University, Giza, 12613, Egypt
| | - Mahmoud Moawad
- Department of Surgical Pathology, National Cancer Institute, Cairo University, Egypt
| | - Wafaa A Ahmed
- Department of Cancer Biology, National Cancer Institute, Cairo University, Egypt
| |
Collapse
|
5
|
Chu S, Ma Y. Evaluation of combination gene therapy with SLC22A18 upregulation and sequence binding protein 1 downregulation for glioma U251 cells in vitro and in vivo. GLIOMA 2020. [DOI: 10.4103/glioma.glioma_19_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
6
|
SATB family chromatin organizers as master regulators of tumor progression. Oncogene 2018; 38:1989-2004. [PMID: 30413763 DOI: 10.1038/s41388-018-0541-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/30/2018] [Accepted: 09/02/2018] [Indexed: 02/07/2023]
Abstract
SATB (Special AT-rich binding protein) family proteins have emerged as key regulators that integrate higher-order chromatin organization with the regulation of gene expression. Studies over the past decade have elucidated the specific roles of SATB1 and SATB2, two closely related members of this family, in cancer progression. SATB family chromatin organizers play diverse and important roles in regulating the dynamic equilibrium of apoptosis, cell invasion, metastasis, proliferation, angiogenesis, and immune modulation. This review highlights cellular and molecular events governed by SATB1 influencing the structural organization of chromatin and interacting with several co-activators and co-repressors of transcription towards tumor progression. SATB1 expression across tumor cell types generates cellular and molecular heterogeneity culminating in tumor relapse and metastasis. SATB1 exhibits dynamic expression within intratumoral cell types regulated by the tumor microenvironment, which culminates towards tumor progression. Recent studies suggested that cell-specific expression of SATB1 across tumor recruited dendritic cells (DC), cytotoxic T lymphocytes (CTL), T regulatory cells (Tregs) and tumor epithelial cells along with tumor microenvironment act as primary determinants of tumor progression and tumor inflammation. In contrast, SATB2 is differentially expressed in an array of cancer types and is involved in tumorigenesis. Survival analysis for patients across an array of cancer types correlated with expression of SATB family chromatin organizers suggested tissue-specific expression of SATB1 and SATB2 contributing to disease prognosis. In this context, it is pertinent to understand molecular players, cellular pathways, genetic and epigenetic mechanisms governed by cell types within tumors regulated by SATB proteins. We propose that patient survival analysis based on the expression profile of SATB chromatin organizers would facilitate their unequivocal establishment as prognostic markers and therapeutic targets for cancer therapy.
Collapse
|
7
|
Yang B, Ma YB, Chu SH. Silencing SATB1 overcomes temozolomide resistance by downregulating MGMT expression and upregulating SLC22A18 expression in human glioblastoma cells. Cancer Gene Ther 2018; 25:309-316. [PMID: 30140041 DOI: 10.1038/s41417-018-0040-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 07/02/2018] [Accepted: 07/07/2018] [Indexed: 12/26/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common malignant tumor of the central nervous system and has a very poor prognosis. Currently, patients were treated by resection followed by radiotherapy plus concurrent temozolomide (TMZ) chemotherapy. However, many patients are resistant to TMZ-induced DNA damage because of upregulated expression of the DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT). In this study, upregulation of SATB1 and MGMT, and downregulation of SLC22A18 resulted in acquisition of TMZ resistance in GBM U87 cells. Inactivation of special AT-rich sequence-binding protein 1 (SATB1) using short hairpin RNA (shRNA) downregulated MGMT expression and upregulated solute carrier family 22 member 18 (SLC22A18) expression in GBM cells. This suggested SATB1-mediated posttranscriptional regulation of the MGMT and SLC22A18 protein levels. Immunohistochemical analysis of malignant glioma specimens demonstrated a significant positive correlation between the levels of MGMT and SATB1, and a negative correlation between the levels of SLC22A18 and SATB1. Importantly, in recurrent, compared with the primary, lesions in 15 paired identical tumors, the SATB1 and MGMT protein levels were increased and the SLC22A18 levels were decreased. Finally, in TMZ-resistant GBM, SATB1 knockdown enhanced TMZ efficacy. Consequently, SATB1 inhibition might be a promising strategy combined with TMZ chemotherapy to treat TMZ-resistant GBM.
Collapse
Affiliation(s)
- Biao Yang
- Department of Neurosurgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China
| | - Yan-Bin Ma
- Department of Neurosurgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China
| | - Sheng-Hua Chu
- Department of Neurosurgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China.
| |
Collapse
|
8
|
Functional relevance of SATB1 in immune regulation and tumorigenesis. Biomed Pharmacother 2018; 104:87-93. [DOI: 10.1016/j.biopha.2018.05.045] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/05/2018] [Accepted: 05/08/2018] [Indexed: 02/07/2023] Open
|
9
|
The Special AT-rich Sequence Binding Protein 1 (SATB1) and its role in solid tumors. Cancer Lett 2018; 417:96-111. [DOI: 10.1016/j.canlet.2017.12.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/19/2017] [Accepted: 12/21/2017] [Indexed: 02/07/2023]
|
10
|
Aigner A, Kögel D. Nanoparticle/siRNA-based therapy strategies in glioma: which nanoparticles, which siRNAs? Nanomedicine (Lond) 2017; 13:89-103. [PMID: 29199893 DOI: 10.2217/nnm-2017-0230] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Nanomedicines allow for the delivery of small interfering RNAs (siRNAs) that are otherwise barely suitable as therapeutics for inducing RNA interference (RNAi). In preclinical studies on siRNA-based glioma treatment in vivo, various groups of nanoparticle systems, routes of administration and target genes have been explored. Targeted delivery by functionalization of nanoparticles with a ligand for crossing the blood-brain barrier and/or for enhanced target cell transfection has been described as well. Focusing on nanoparticle developments in the last approximately 10 years, this review article gives a comprehensive overview of nanoparticle systems for siRNA delivery into glioma and of preclinical in vivo studies. Furthermore, it discusses various target genes and highlights promising strategies with regard to target gene selection and combination therapies.
Collapse
Affiliation(s)
- Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology & Toxicology, Clinical Pharmacology, University of Leipzig, Germany
| | - Donat Kögel
- Experimental Neurosurgery, Neuroscience Center, Goethe University Hospital, Frankfurt am Main, Germany
| |
Collapse
|
11
|
Malhotra M, Toulouse A, Godinho BMDC, Mc Carthy DJ, Cryan JF, O'Driscoll CM. RNAi therapeutics for brain cancer: current advancements in RNAi delivery strategies. MOLECULAR BIOSYSTEMS 2016; 11:2635-57. [PMID: 26135606 DOI: 10.1039/c5mb00278h] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Malignant primary brain tumors are aggressive cancerous cells that invade the surrounding tissues of the central nervous system. The current treatment options for malignant brain tumors are limited due to the inability to cross the blood-brain barrier. The advancements in current research has identified and characterized certain molecular markers that are essential for tumor survival, progression, metastasis and angiogenesis. These molecular markers have served as therapeutic targets for the RNAi based therapies, which enable site-specific silencing of the gene responsible for tumor proliferation. However, to bring about therapeutic success, an efficient delivery carrier that can cross the blood-brain barrier and reach the targeted site is essential. The current review focuses on the potential of targeted, non-viral and viral particles containing RNAi therapeutic molecules as delivery strategies specifically for brain tumors.
Collapse
Affiliation(s)
- Meenakshi Malhotra
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | | | | | | | | | | |
Collapse
|
12
|
Piktel E, Niemirowicz K, Wątek M, Wollny T, Deptuła P, Bucki R. Recent insights in nanotechnology-based drugs and formulations designed for effective anti-cancer therapy. J Nanobiotechnology 2016; 14:39. [PMID: 27229857 PMCID: PMC4881065 DOI: 10.1186/s12951-016-0193-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/17/2016] [Indexed: 12/18/2022] Open
Abstract
The rapid development of nanotechnology provides alternative approaches to overcome several limitations of conventional anti-cancer therapy. Drug targeting using functionalized nanoparticles to advance their transport to the dedicated site, became a new standard in novel anti-cancer methods. In effect, the employment of nanoparticles during design of antineoplastic drugs helps to improve pharmacokinetic properties, with subsequent development of high specific, non-toxic and biocompatible anti-cancer agents. However, the physicochemical and biological diversity of nanomaterials and a broad spectrum of unique features influencing their biological action requires continuous research to assess their activity. Among numerous nanosystems designed to eradicate cancer cells, only a limited number of them entered the clinical trials. It is anticipated that progress in development of nanotechnology-based anti-cancer materials will provide modern, individualized anti-cancer therapies assuring decrease in morbidity and mortality from cancer diseases. In this review we discussed the implication of nanomaterials in design of new drugs for effective antineoplastic therapy and describe a variety of mechanisms and challenges for selective tumor targeting. We emphasized the recent advantages in the field of nanotechnology-based strategies to fight cancer and discussed their part in effective anti-cancer therapy and successful drug delivery.
Collapse
Affiliation(s)
- Ewelina Piktel
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2c, 15-222, Bialystok, Poland
| | - Katarzyna Niemirowicz
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2c, 15-222, Bialystok, Poland
| | - Marzena Wątek
- Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-317, Kielce, Poland
| | - Tomasz Wollny
- Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-317, Kielce, Poland
| | - Piotr Deptuła
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2c, 15-222, Bialystok, Poland
| | - Robert Bucki
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2c, 15-222, Bialystok, Poland. .,Department of Physiology, Pathophysiology and Immunology of Infections, The Faculty of Health Sciences of the Jan Kochanowski University in Kielce, Kielce, Al. IX Wieków Kielc 19, 25-317, Kielce, Poland.
| |
Collapse
|
13
|
Oltolina F, Gregoletto L, Colangelo D, Gómez-Morales J, Delgado-López JM, Prat M. Monoclonal antibody-targeted fluorescein-5-isothiocyanate-labeled biomimetic nanoapatites: a promising fluorescent probe for imaging applications. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2015; 31:1766-1775. [PMID: 25602940 DOI: 10.1021/la503747s] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Multifunctional biomimetic nanoparticles (NPs) are acquiring increasing interest as carriers in medicine and basic research since they can efficiently combine labels for subsequent tracking, moieties for specific cell targeting, and bioactive molecules, e.g., drugs. In particular, because of their easy synthesis, low cost, good biocompatibility, high resorbability, easy surface functionalization, and pH-dependent solubility, nanocrystalline apatites are promising candidates as nanocarriers. This work describes the synthesis and characterization of bioinspired apatite nanoparticles to be used as fluorescent nanocarriers targeted against the Met/hepatocyte growth factor receptor, which is considered a tumor associated cell surface marker of many cancers. To this aim the nanoparticles have been labeled with Fluorescein-5-isothiocyanate (FITC) by simple isothermal adsorption, in the absence of organic, possibly toxic, molecules, and then functionalized with a monoclonal antibody (mAb) directed against such a receptor. Direct labeling of the nanoparticles allowed tracking the moieties with spatiotemporal resolution and thus following their interaction with cells, expressing or not the targeted receptor, as well as their fate in vitro. Cytofluorometry and confocal microscopy experiments showed that the functionalized nanocarriers, which emitted a strong fluorescent signal, were rapidly and specifically internalized in cells expressing the receptor. Indeed, we found that, once inside the cells expressing the receptor, mAb-functionalized FITC nanoparticles partially dissociated in their two components, with some mAbs being recycled to the cell surface and the FITC-labeled nanoparticles remaining in the cytosol. This work thus shows that FITC-labeled nanoapatites are very promising probes for targeted cell imaging applications.
Collapse
Affiliation(s)
- Francesca Oltolina
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale "Amedeo Avogadro" , Via Solaroli 17, 28100 Novara, Italy
| | | | | | | | | | | |
Collapse
|
14
|
Nicotinamide induces apoptosis of F9 mouse teratocarcinoma stem cells by downregulation of SATB1 expression. Tumour Biol 2015; 36:4339-48. [PMID: 25596087 DOI: 10.1007/s13277-015-3073-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 01/08/2015] [Indexed: 10/24/2022] Open
Abstract
The aim of this study was to decide whether nicotinamide (NA) could induce apoptosis of F9 mouse teratocarcinoma stem cells (MF9) by downregulation of special AT-rich sequence binding protein 1 (SATB1) expression. We used different concentrations of NA (0, 1.5, 2, and 2.5 mmol/L) to treat MF9 cells and analyze SATB1 expression by RT-qPCR and Western blotting; in addition, the cell proliferation was detected in a microplate reader with Cell Counting Kit-8 (CCK-8), and the cell cycle and apoptosis were analyzed using flow cytometry. We found that the expression of SATB1 was decreased significantly in NA-treated groups than in the control group, and its expression level was inversely related to the NA concentration. In addition, CCK-8 analysis showed that NA significantly inhibited the proliferation of MF9 cells, and flow cytometry showed that NA blocked MF9 cells to G1 phase and significantly promoted apoptosis in any treated groups. To confirm the results, we constructed small interference RNA (siRNA) targeting at mouse SATB1 and transferred into MF9 cells. The results indicated that the expression of SATB1 in both mRNA and protein levels was significantly decreased after cells transferred with siRNA sequence for 48 h, the proliferation of MF9 cells was significantly inhibited, and most of MF9 cells were blocked at G1 phase, and the apoptosis rate was increased obviously. The results showed that NA could inhibit the proliferation and induce apoptosis of MF9 cells. These findings might be used as an efficient candidate for teratocarcinoma therapy.
Collapse
|