1
|
Sun C, Gui J, Sheng Y, Huang L, Zhu X, Huang K. Specific signaling pathways mediated programmed cell death in tumor microenvironment and target therapies. Discov Oncol 2025; 16:776. [PMID: 40377777 PMCID: PMC12084487 DOI: 10.1007/s12672-025-02592-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
Increasing evidence has shown that programmed cell death (PCD) plays a crucial role in tumorigenesis and cancer progression. The components of PCD are complex and include various mechanisms such as apoptosis, necroptosis, alkaliptosis, oxeiptosis, and anoikis, all of which are interrelated in their functions and regulatory pathways. Given the significance of these processes, it is essential to conduct a comprehensive study on PCD to elucidate its multifaceted nature. Key signaling pathways, particularly the caspase signaling pathway, the RIPK1/RIPK3/MLKL pathway, and the mTOR signaling pathway, are pivotal in regulating PCD and influencing tumor progression. In this review, we briefly describe the generation mechanisms of different PCD components and focus on the regulatory mechanisms of these three major signaling pathways within the context of global PCD. Furthermore, we discuss various tumor therapeutic compounds that target different signaling axes of these pathways, which may provide novel strategies for effective tumor therapy and help improve patient outcomes in cancer treatment.
Collapse
Affiliation(s)
- Chengpeng Sun
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Jiawei Gui
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Yilei Sheng
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Le Huang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China
| | - Xingen Zhu
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Nanchang, 330006, Jiangxi, China.
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| | - Kai Huang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Nanchang, 330006, Jiangxi, China.
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
2
|
Jaksic Karisik M, Jović Orsini N, Carkic J, Lazarevic M, Mitić D, Jokanovic B, Jokanović V, Milasin J. A Carbon-Based Nanomaterial with Dichotomous Effects: Antineoplastic on Oral Cancer Cells and Osteoinductive/Chondroinductive on Dental Pulp Stem Cells. J Funct Biomater 2025; 16:109. [PMID: 40137387 PMCID: PMC11943258 DOI: 10.3390/jfb16030109] [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: 12/18/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Oral cancer is an aggressive malignancy with modest survival rates. It also causes disfigurement following surgical removal of the tumor, thus highlighting the need for new cancer treatment and tissue repair modalities. Carbon-based nanomaterials have emerged as promising tools in both anticancer and regenerative therapies. OBJECTIVES We aimed to synthesize a new carbon-based nanomaterial (CBN) and test its antineoplastic effects, as well as its potential regenerative capacity. MATERIALS AND METHODS A carbon nanomaterial, obtained by ball milling graphite flakes, was functionalized with polyvinylpyrrolidone (CBN/PVP). Its physicochemical properties were explored with X-ray diffraction (XRD), attenuated total reflection-Fourier transform infrared spectroscopy (ATR-FTIR), micro-Raman spectroscopy, fluorescent and scanning electron microscopy, and wettability analysis. For the antineoplastic effects investigation, oral cancer cells were treated with CBN/PVP and examined with MTT and migration assays, as well as cell-cycle and ROS production analyses. Gene expression was determined by qPCR. To examine the pro-regenerative capacity of CBN/PVP, dental pulp stem cell cultures (DPSCs) were treated with the nanomaterial and subjected to osteo- and chondro-induction. RESULTS Lower concentrations of CBN/PVP (50, 100 μg/mL) applied on cancer cells exerted remarkable cytotoxic effects, induced G1 cell-cycle arrest, and reduced cancer cell invasion potential by different mechanisms, including downregulation of the PI3K/AKT/mTOR pathway. In contrast, the addition of 50 µg/mL of CBN/PVP to DPSCs stimulated their survival and proliferation. CBN/PVP significantly enhanced both the osteogenic (p < 0.05) and chondrogenic (p < 0.01) induction of DPSCs. CONCLUSIONS The novel carbon-based nanomaterial displays unique characteristics, making it suitable in anticancer and regenerative therapies concomitantly.
Collapse
Affiliation(s)
- Milica Jaksic Karisik
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.J.K.); (J.C.); (M.L.); (D.M.)
| | - Nataša Jović Orsini
- Vinča Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11001 Belgrade, Serbia;
| | - Jelena Carkic
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.J.K.); (J.C.); (M.L.); (D.M.)
| | - Milos Lazarevic
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.J.K.); (J.C.); (M.L.); (D.M.)
| | - Dijana Mitić
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.J.K.); (J.C.); (M.L.); (D.M.)
| | | | | | - Jelena Milasin
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.J.K.); (J.C.); (M.L.); (D.M.)
| |
Collapse
|
3
|
Li R, He T, Yang M, Xu J, Li Y, Wang X, Guo X, Li M, Xu L. Regulation of Bacillus Calmette-Guérin-induced macrophage autophagy and apoptosis by the AMPK-mTOR-ULK1 pathway. Microbiol Res 2025; 290:127952. [PMID: 39476518 DOI: 10.1016/j.micres.2024.127952] [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: 05/09/2024] [Revised: 08/02/2024] [Accepted: 10/24/2024] [Indexed: 12/12/2024]
Abstract
Tuberculosis (TB) is a chronic wasting infectious disease caused by Mycobacterium tuberculosis (MTB) or Mycobacterium bovis that can be transmitted among people and domestic animals. During the development of TB, macrophages of the innate immune system can act against MTB via autophagy and apoptosis to prevent the spread of the disease. Among the many autophagy regulatory pathways, the adenosine monophosphate (AMP)-activated protein kinase (AMPK)-mammalian rapamycin target protein (mTOR)-Unc-51-like kinase 1 (ULK1) pathway has received considerable attention. This study investigates the regulatory role of the AMPK-mTOR-ULK1 pathway in attenuating M. bovis Bacillus Calmette-Guérin (BCG)-induced autophagy and apoptosis in murine monocyte macrophages (RAW264.7). Changes in macrophage autophagy and apoptosis were analyzed using the AMPK activator AICAR and inhibitor Compound C to interfere with the AMPK-mTOR-ULK1 pathway and siRNA to silence the pathway. Consequently, BCG stimulation of macrophages significantly activated the AMPK-mTOR-ULK1 pathway while BCG-induced macrophage AMPK activation promoted macrophage autophagy and apoptosis. Activation of the AMPK-mTOR-ULK1 pathway by AICAR significantly improved autophagy occurrence in BCG-induced macrophages and increased apoptosis while Compound C with siRNA produced opposing effects by attenuating autophagy and apoptosis in BCG-induced macrophages. Thus, the AMPK-mTOR-ULK1 pathway has a dual regulatory role in BCG-induced macrophage autophagy and apoptosis and may have synergistic effects. This study analyzes the mechanism of resistance of host cells to MTB and provides a theoretical basis for new therapeutic strategies and related drug development.
Collapse
Affiliation(s)
- Ruiqian Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Tianle He
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Min Yang
- Guyuan Vocational and Technical School, Guyuan, Ningxia 756000, China
| | - Jinghua Xu
- COFCO Feed (Yinchuan) Co., Ltd., Lingwu, Ningxia 750499, China
| | - Yongqin Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Xueyan Wang
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Xuelian Guo
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Mingzhu Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Lihua Xu
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China.
| |
Collapse
|
4
|
Teymourlouei AS, Naghib SM, Mozafari MR. Stimuli-responsive Graphene-polysaccharide Nanocomposites for Drug Delivery and Tissue Engineering. Curr Org Synth 2025; 22:211-233. [PMID: 39962959 DOI: 10.2174/0115701794298435240324175513] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/19/2024] [Accepted: 03/08/2024] [Indexed: 05/10/2025]
Abstract
Natural polysaccharide-based nanoparticles are known for their non-toxic nature and diverse medical applications. Graphene oxide (GO) nanoparticles show potential in cancer treatment due to their ability to target medication delivery and influence ROS generation. These nanocomposites are versatile in gene transport, therapy, and photodynamic therapy, especially when surface-modified. Proper dispersion and functionalization of GO in polymer matrices are crucial, with examples like hyaluronic acid-functionalized GO offering versatile platforms for cancer drug administration. The potential of graphene oxide extends to cancer phototherapy, electronic nanowires, hydrogels, antibacterial nanocomposites, and environmental applications. When activated by polysaccharides, graphene-based nanocomposites exhibit anti-inflammatory and anticancer properties, making them valuable across various industries, including water treatment.
Collapse
Affiliation(s)
- Arman Seifallahi Teymourlouei
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, 1684613114, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, 1684613114, Iran
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC, 3168, Australia
| |
Collapse
|
5
|
Zhou X, Zhang Q, Zhu H, Ouyang G, Wang X, Cai Y. High Carbonyl Graphene Oxide Suppresses Colorectal Cancer Cell Proliferation and Migration by Inducing Ferroptosis via the System Xc-/GSH/GPX4 Axis. Pharmaceutics 2024; 16:1605. [PMID: 39771583 PMCID: PMC11678421 DOI: 10.3390/pharmaceutics16121605] [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: 11/07/2024] [Revised: 11/25/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Colorectal cancer (CRC) is characterized by a high rate of both incidence and mortality, and its treatment outcomes are often affected by recurrence and drug resistance. Ferroptosis, an iron-dependent programmed cell death mechanism triggered by lipid peroxidation, has recently gained attention as a potential therapeutic target. Graphene oxide (GO), known for its oxygen-containing functional groups, biocompatibility, and potential for functionalization, holds promise in cancer treatment. However, its role in ferroptosis induction in CRC remains underexplored. The objective of this study was to investigate the effects of High Carbonyl Graphene Oxide (HC-GO) on ferroptosis in CRC and elucidate the underlying mechanisms. METHODS In vitro assays were conducted to evaluate the impact of HC-GO on CRC cell proliferation, mitochondrial function, iron accumulation, lipid peroxidation, and reactive oxygen species (ROS) production. The ferroptosis inhibitor Fer-1 was used to confirm the role of ferroptosis in HC-GO's anti-tumor effects. In vivo, the anti-tumor activity of HC-GO was assessed in a CRC xenograft model, with organ toxicity evaluated. RESULTS HC-GO significantly inhibited CRC cell proliferation, induced mitochondrial damage, and enhanced iron accumulation, lipid peroxidation, and ROS production. It also downregulated the ferroptosis-inhibiting proteins GPX4 and SLC7A11, which were reversed by Fer-1, confirming the involvement of ferroptosis in HC-GO's anti-cancer effects. In vivo, HC-GO significantly suppressed tumor growth without noticeable toxicity to vital organs. CONCLUSIONS HC-GO triggered ferroptosis in CRC cells by suppressing the System Xc-/GSH/GPX4 pathway, providing a novel therapeutic strategy for CRC treatment. These findings suggest HC-GO as a promising nanomedicine for clinical application, warranting further investigation to explore its potential in CRC therapy.
Collapse
Affiliation(s)
- Xiecheng Zhou
- Department of General Surgery, The Fifth People’s Hospital of Shanghai, Fudan University, Shanghai 200240, China; (X.Z.); (H.Z.)
- Center of Community-Based Health Research, Fudan University, Shanghai 200240, China;
| | - Qixing Zhang
- Center of Community-Based Health Research, Fudan University, Shanghai 200240, China;
- Department of Pediatrics, The Fifth People’s Hospital of Shanghai, Fudan University, Shanghai 200240, China
| | - Haoran Zhu
- Department of General Surgery, The Fifth People’s Hospital of Shanghai, Fudan University, Shanghai 200240, China; (X.Z.); (H.Z.)
- Center of Community-Based Health Research, Fudan University, Shanghai 200240, China;
| | - Guangxiong Ouyang
- Department of General Surgery, The Fifth People’s Hospital of Shanghai, Fudan University, Shanghai 200240, China; (X.Z.); (H.Z.)
- Center of Community-Based Health Research, Fudan University, Shanghai 200240, China;
| | - Xin Wang
- Department of General Surgery, The Fifth People’s Hospital of Shanghai, Fudan University, Shanghai 200240, China; (X.Z.); (H.Z.)
- Center of Community-Based Health Research, Fudan University, Shanghai 200240, China;
| | - Yuankun Cai
- Department of General Surgery, The Fifth People’s Hospital of Shanghai, Fudan University, Shanghai 200240, China; (X.Z.); (H.Z.)
- Center of Community-Based Health Research, Fudan University, Shanghai 200240, China;
| |
Collapse
|
6
|
Flasz B, Babczyńska A, Tarnawska M, Ajay AK, Kędziorski A, Napora-Rutkowski Ł, Augustyniak M. Graphene oxide in low concentrations can change mitochondrial potential, autophagy, and apoptosis paths in two strains of invertebrates with different life strategies. Biochem Biophys Res Commun 2024; 736:150898. [PMID: 39467354 DOI: 10.1016/j.bbrc.2024.150898] [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: 09/17/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 10/30/2024]
Abstract
Nanoparticles, like graphene oxide (GO), are particles with unique physiochemical properties that enable their wide application in various areas of life. The effects of GO on individual cell organelles like mitochondria and the effects of interactions are worth investigating, as they can activate multiple cellular processes, such as autophagy or apoptosis. Mitochondrial injury plays an essential role in the majority of cell death routines. In the project, we investigated cell health status measured as mitochondrial inner membrane depolarization, autophagy, and apoptosis induction during long-term GO administration in food (0.02 μg g-1 and 0.2 μg g-1 of food). Two unique Acheta domesticus strains that differ in life strategy were used: wild-type and long-lived at three different life stages (larva, young adult, mature adult). The changes in mitochondrial trans-membrane potential were marked in the wild-type strain. The autophagy was lower in all GO-treated groups in both strains, and the apoptosis was lower in both strains in the mature adult crickets. Low GO concentrations treatment for the whole life, despite mitochondrial dysfunction, may lead to inhibition of autophagy and apoptosis by arresting the cell cycle for the duration of repair, and other repair tools are involved in the process of restoring homeostasis.
Collapse
Affiliation(s)
- Barbara Flasz
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Poland.
| | - Agnieszka Babczyńska
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Poland
| | - Monika Tarnawska
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Poland
| | - Amrendra K Ajay
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Andrzej Kędziorski
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Poland
| | - Łukasz Napora-Rutkowski
- Polish Academy of Sciences, Institute of Ichthyobiology and Aquaculture in Gołysz, 43-520, Chybie, Poland
| | - Maria Augustyniak
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Poland
| |
Collapse
|
7
|
Tamtaji OR, Ostadian A, Homayoonfal M, Nejati M, Mahjoubin-Tehran M, Nabavizadeh F, Ghelichi E, Mohammadzadeh B, Karimi M, Rahimian N, Mirzaei H. Cerium(IV) oxide:silver/graphene oxide (CeO2:Ag/GO) nanoparticles modulate gene expression and inhibit colorectal cancer cell growth: a pathway-centric therapeutic approach. Cancer Nanotechnol 2024; 15:62. [DOI: 10.1186/s12645-024-00300-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/18/2024] [Indexed: 01/06/2025] Open
|
8
|
Luobin L, Wanxin H, Yingxin G, Qinzhou Z, Zefeng L, Danyang W, Huaqin L. Nanomedicine-induced programmed cell death in cancer therapy: mechanisms and perspectives. Cell Death Discov 2024; 10:386. [PMID: 39209834 PMCID: PMC11362291 DOI: 10.1038/s41420-024-02121-0] [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: 05/16/2024] [Revised: 07/20/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The balance of programmed cell death (PCD) mechanisms, including apoptosis, autophagy, necroptosis and others, is pivotal in cancer progression and treatment. Dysregulation of these pathways results in uncontrolled cell growth and resistance to conventional therapies. Nanomedicine offers a promising solution in oncology through targeted drug delivery enabling precise targeting of cancer cells while preserving healthy tissues. This approach reduces the side effects of traditional chemotherapy and enhances treatment efficacy by engaging PCD pathways. We details each PCD pathway, their mechanisms, and innovative nanomedicine strategies to activate these pathways, thereby enhancing therapeutic specificity and minimizing harm to healthy tissues. The precision of nanotechnology in targeting PCD pathways promises significant improvements in cancer treatment outcomes. This synergy between nanotechnology and targeted PCD activation could lead to more effective and less toxic cancer therapies, heralding a new era in cancer treatment.
Collapse
Affiliation(s)
- Lin Luobin
- School of Health Sciences, Guangzhou Xinhua University, 19 Huamei Road, Tianhe District, Guangzhou, 510520, China
- School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - He Wanxin
- School of Health Sciences, Guangzhou Xinhua University, 19 Huamei Road, Tianhe District, Guangzhou, 510520, China
| | - Guo Yingxin
- School of Health Sciences, Guangzhou Xinhua University, 19 Huamei Road, Tianhe District, Guangzhou, 510520, China
| | - Zheng Qinzhou
- School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Liang Zefeng
- School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Wu Danyang
- School of Health Sciences, Guangzhou Xinhua University, 19 Huamei Road, Tianhe District, Guangzhou, 510520, China
| | - Li Huaqin
- School of Health Sciences, Guangzhou Xinhua University, 19 Huamei Road, Tianhe District, Guangzhou, 510520, China.
| |
Collapse
|
9
|
Batool M, Qazi REM, Mudassir MA, Sajid Z, Zaman R, Rauf MA, Kousar S, Ahmad I, Rehman FU, Mian AA. Titania-Graphene Oxide Nanocomposite-Based Philadelphia-Positive Leukemia Therapy. ACS APPLIED BIO MATERIALS 2024; 7:4352-4365. [PMID: 38900491 DOI: 10.1021/acsabm.4c00207] [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] [Indexed: 06/21/2024]
Abstract
Philadelphia-positive (Ph+) leukemia is a type of blood cancer also known as acute lymphoblastic leukemia (ALL), affecting 20-30% of adults diagnosed worldwide and having an engraved prognosis as compared to other types of leukemia. The current treatment regimens mainly rely on tyrosine kinase inhibitors (TKIs) and bone marrow transplants. To date, several generations of TKIs have been developed due to associated resistance and frequent relapse, with cardiovascular system anomalies being the most devastating complication. Nanotechnology has the potential to address these limitations by the targeted drug delivery and controlled release of TKIs. This study focused on the titanium dioxide (TiO2) and graphene oxide (GO) nanocomposite employment to load nilotinib and ponatinib TKIs for therapy of Ph+ leukemia cell line (K562) and Ba/F3 cells engineered to express BCR-ABL oncogene. Meanwhile, after treatment, the oncogene expressing fibroblast cells (Rat-1 P185) were evaluated for their colony formation ability under 3D conditions. To validate the nanocomposite formation, the TiO2-GO nanocomposites were characterized by scanning electron microscope, DLS, XRD, FTIR, zeta potential, EDX, and element mapping. The TKI-loaded TiO2-GO was not inferior to the free drugs after evaluating their effects by a cell viability assay (XTT), apoptosis induction, and colony formation inhibition. The cell signaling pathways of the mammalian target of rapamycin (mTOR), signal transducers and activators of transcription 5 (STAT5), and extracellular signal-regulated kinase (Erk1/2) were also investigated by Western blot. These signaling pathways were significantly downregulated in the TKI-loaded TiO2-GO-treated groups. Based on the findings above, we can conclude that TiO2-GO exhibited excellent drug delivery potential that can be used for Ph+ leukemia therapy in the future, subject to further investigations.
Collapse
MESH Headings
- Graphite/chemistry
- Graphite/pharmacology
- Titanium/chemistry
- Titanium/pharmacology
- Nanocomposites/chemistry
- Humans
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Cell Survival/drug effects
- Materials Testing
- Particle Size
- Drug Screening Assays, Antitumor
- Biocompatible Materials/chemistry
- Biocompatible Materials/pharmacology
- Cell Proliferation/drug effects
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/chemistry
- Animals
Collapse
Affiliation(s)
- Maria Batool
- Institute of Chemistry, Khwaja Fareed University of Engineering and Information Technology (KFUEIT), Rahim Yar Khan 64200, Pakistan
| | - Rida-E-Maria Qazi
- Centre for Regenerative Medicine and Stem Cells Research, First Flour, Juma Building, Aga Khan University, Stadium Road, Karachi 74800, Sindh, Pakistan
| | - Muhammad Ahmad Mudassir
- Institute of Chemistry, Khwaja Fareed University of Engineering and Information Technology (KFUEIT), Rahim Yar Khan 64200, Pakistan
- Chemistry Department, University of Management and Technology (UMT), Sialkot Campus, Sialkot 51310, Pakistan
| | - Zahra Sajid
- Centre for Regenerative Medicine and Stem Cells Research, First Flour, Juma Building, Aga Khan University, Stadium Road, Karachi 74800, Sindh, Pakistan
| | - Rena Zaman
- Centre for Regenerative Medicine and Stem Cells Research, First Flour, Juma Building, Aga Khan University, Stadium Road, Karachi 74800, Sindh, Pakistan
| | - Mhd Ahmar Rauf
- Rogel Cancer Center, Department of Internal Medicine, Heme Oncology Unit, University of Michigan, Ann Arbor 48109-1382, United States
| | - Shazia Kousar
- Institute of Chemistry, Khwaja Fareed University of Engineering and Information Technology (KFUEIT), Rahim Yar Khan 64200, Pakistan
| | - Israr Ahmad
- Institute of Chemistry, Khwaja Fareed University of Engineering and Information Technology (KFUEIT), Rahim Yar Khan 64200, Pakistan
| | - Fawad Ur Rehman
- Centre for Regenerative Medicine and Stem Cells Research, First Flour, Juma Building, Aga Khan University, Stadium Road, Karachi 74800, Sindh, Pakistan
| | - Afsar Ali Mian
- Centre for Regenerative Medicine and Stem Cells Research, First Flour, Juma Building, Aga Khan University, Stadium Road, Karachi 74800, Sindh, Pakistan
| |
Collapse
|
10
|
Zhao G, Wang Y, Fan Z, Xiong J, Ertas YN, Ashammakhi N, Wang J, Ma T. Nanomaterials in crossroad of autophagy control in human cancers: Amplification of cell death mechanisms. Cancer Lett 2024; 591:216860. [PMID: 38583650 DOI: 10.1016/j.canlet.2024.216860] [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: 01/30/2024] [Revised: 03/24/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
Cancer is the result of genetic abnormalities that cause normal cells to grow into neoplastic cells. Cancer is characterized by several distinct features, such as uncontrolled cell growth, extensive spreading to other parts of the body, and the ability to resist treatment. The scientists have stressed the development of nanostructures as novel therapeutic options in suppressing cancer, in response to the emergence of resistance to standard medicines. One of the specific mechanisms with dysregulation during cancer is autophagy. Nanomaterials have the ability to specifically carry medications and genes, and they can also enhance the responsiveness of tumor cells to standard therapy while promoting drug sensitivity. The primary mechanism in this process relies on autophagosomes and their fusion with lysosomes to break down the components of the cytoplasm. While autophagy was initially described as a form of cellular demise, it has been demonstrated to play a crucial role in controlling metastasis, proliferation, and treatment resistance in human malignancies. The pharmacokinetic profile of autophagy modulators is poor, despite their development for use in cancer therapy. Consequently, nanoparticles have been developed for the purpose of delivering medications and autophagy modulators selectively and specifically to the cancer process. Furthermore, several categories of nanoparticles have demonstrated the ability to regulate autophagy, which plays a crucial role in defining the biological characteristics and response to therapy of tumor cells.
Collapse
Affiliation(s)
- Gang Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yutao Wang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing, 100000, China
| | - Zhongru Fan
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Jian Xiong
- Department of Obstetrics and Gynaecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yavuz Nuri Ertas
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, 38039, Türkiye; Department of Biomedical Engineering, Erciyes University, Kayseri, 39039, Türkiye.
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ), Department of Biomedical Engineering, College of Engineering and Human Medicine, Michigan State University, East Lansing, MI, 48824, USA.
| | - Jianfeng Wang
- Department of Urology, First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Ting Ma
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
11
|
Uzdrowska K, Knap N, Gulczynski J, Kuban-Jankowska A, Struck-Lewicka W, Markuszewski MJ, Bączek T, Izycka-Swieszewska E, Gorska-Ponikowska M. Chasing Graphene-Based Anticancer Drugs: Where are We Now on the Biomedical Graphene Roadmap? Int J Nanomedicine 2024; 19:3973-3989. [PMID: 38711615 PMCID: PMC11073537 DOI: 10.2147/ijn.s447397] [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: 10/31/2023] [Accepted: 03/28/2024] [Indexed: 05/08/2024] Open
Abstract
Graphene and graphene-based materials have attracted growing interest for potential applications in medicine because of their good biocompatibility, cargo capability and possible surface functionalizations. In parallel, prototypic graphene-based devices have been developed to diagnose, imaging and track tumor growth in cancer patients. There is a growing number of reports on the use of graphene and its functionalized derivatives in the design of innovative drugs delivery systems, photothermal and photodynamic cancer therapy, and as a platform to combine multiple therapies. The aim of this review is to introduce the latest scientific achievements in the field of innovative composite graphene materials as potentially applied in cancer therapy. The "Technology and Innovation Roadmap" published in the Graphene Flagship indicates, that the first anti-cancer drugs using graphene and graphene-derived materials will have appeared on the market by 2030. However, it is necessary to broaden understanding of graphene-based material interactions with cellular metabolism and signaling at the functional level, as well as toxicity. The main aspects of further research should elucidate how treatment methods (e.g., photothermal therapy, photodynamic therapy, combination therapy) and the physicochemical properties of graphene materials influence their ability to modulate autophagy and kill cancer cells. Interestingly, recent scientific reports also prove that graphene nanocomposites modulate cancer cell death by inducing precise autophagy dysfunctions caused by lysosome damage. It turns out as well that developing photothermal oncological treatments, it should be taken into account that near-infrared-II radiation (1000-1500 nm) is a better option than NIR-I (750-1000 nm) because it can penetrate deeper into tissues due to less scattering at longer wavelengths radiation.
Collapse
Affiliation(s)
- Katarzyna Uzdrowska
- Department of Medical Chemistry, Medical University of Gdansk, Gdansk, 80-211, Poland
| | - Narcyz Knap
- Department of Medical Chemistry, Medical University of Gdansk, Gdansk, 80-211, Poland
| | - Jacek Gulczynski
- Faculty of Health Sciences with the Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdansk, 80-211, Poland
| | | | | | | | - Tomasz Bączek
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, 80-416, Poland
| | - Ewa Izycka-Swieszewska
- Faculty of Health Sciences with the Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdansk, 80-211, Poland
| | | |
Collapse
|
12
|
Shen W, Zeng X, Zeng X, Hu B, Ren C, Lin Z, Zhang L, Rui G, Yasen M, Chen X. Trifluoperazine activates AMPK / mTOR / ULK1 signaling pathway to induce mitophagy in osteosarcoma cells. Chem Biol Interact 2024; 392:110904. [PMID: 38360085 DOI: 10.1016/j.cbi.2024.110904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/26/2024] [Accepted: 02/06/2024] [Indexed: 02/17/2024]
Abstract
Osteosarcoma is a prevalent kind of primary bone malignancy. Trifluoperazine, as an antipsychotic drug, has anti-tumor activity against a variety of cancers. Nevertheless, the impact of trifluoperazine on osteosarcoma is unclear. Our investigation aimed to explore the mechanism of trifluoperazine's effect on osteosarcoma. We found that trifluoperazine inhibited 143B and U2-OS osteosarcoma cell proliferation in a method based on the dose. Furthermore, it was shown that trifluoperazine induced the accumulation of reactive oxygen species (ROS) to cause mitochondrial damage and induced mitophagy in osteosarcoma cells. Finally, combined with RNA-seq results, we first demonstrated the AMPK/mTOR/ULK1 signaling pathway as a potential mechanism of trifluoperazine-mediated mitophagy in osteosarcoma cells and can be suppressed by AMPK inhibitor Compound C.
Collapse
Affiliation(s)
- Wenhui Shen
- Department of Orthopedic Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361000, China
| | - Xiangchen Zeng
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Xianhui Zeng
- Department of Infectious Diseases, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, 570206, China
| | - Baoshan Hu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, 361001, China
| | - Chong Ren
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Zhiming Lin
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, 361001, China
| | - Long Zhang
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Gang Rui
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, 361001, China
| | - Miersalijiang Yasen
- Department of Orthopedic Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361000, China.
| | - Xiaohui Chen
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, 361001, China.
| |
Collapse
|
13
|
Morotomi-Yano K, Hayami S, Yano KI. Adhesion States Greatly Affect Cellular Susceptibility to Graphene Oxide: Therapeutic Implications for Cancer Metastasis. Int J Mol Sci 2024; 25:1927. [PMID: 38339205 PMCID: PMC10855874 DOI: 10.3390/ijms25031927] [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: 10/26/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Graphene oxide (GO) has received increasing attention in the life sciences because of its potential for various applications. Although GO is generally considered biocompatible, it can negatively impact cell physiology under some circumstances. Here, we demonstrate that the cytotoxicity of GO greatly varies depending on the cell adhesion states. Human HCT-116 cells in a non-adhered state were more susceptible to GO than those in an adherent state. Apoptosis was partially induced by GO in both adhered and non-adhered cells to a similar extent, suggesting that apoptosis induction does not account for the selective effects of GO on non-adhered cells. GO treatment rapidly decreased intracellular ATP levels in non-adhered cells but not in adhered ones, suggesting ATP depletion as the primary cause of GO-induced cell death. Concurrently, autophagy induction, a cellular response for energy homeostasis, was more evident in non-adhered cells than in adhered cells. Collectively, our observations provide novel insights into GO's action with regard to cell adhesion states. Because the elimination of non-adhered cells is important in preventing cancer metastasis, the selective detrimental effects of GO on non-adhered cells suggest its therapeutic potential for use in cancer metastasis.
Collapse
Affiliation(s)
- Keiko Morotomi-Yano
- Institute of Industrial Nanomaterials, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Shinya Hayami
- Institute of Industrial Nanomaterials, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
- Department of Chemistry, Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Ken-ichi Yano
- Institute of Industrial Nanomaterials, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| |
Collapse
|
14
|
He K, Chen M, Liu J, Du S, Ren C, Zhang J. Nanomedicine for cancer targeted therapy with autophagy regulation. Front Immunol 2024; 14:1238827. [PMID: 38239356 PMCID: PMC10794438 DOI: 10.3389/fimmu.2023.1238827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 12/13/2023] [Indexed: 01/22/2024] Open
Abstract
Nanoparticles have unique physical and chemical properties and are currently widely used in disease diagnosis, drug delivery, and new drug development in biomedicine. In recent years, the role of nanomedical technology in cancer treatment has become increasingly obvious. Autophagy is a multi-step degradation process in cells and an important pathway for material and energy recovery. It is closely related to the occurrence and development of cancer. Because nanomaterials are highly targeted and biosafe, they can be used as carriers to deliver autophagy regulators; in addition to their favorable physicochemical properties, nanomaterials can be employed to carry autophagy inhibitors, reducing the breakdown of chemotherapy drugs by cancer cells and thereby enhancing the drug's efficacy. Furthermore, certain nanomaterials can induce autophagy, triggering oxidative stress-mediated autophagy enhancement and cell apoptosis, thus constraining the progression of cancer cells.There are various types of nanoparticles, including liposomes, micelles, polymers, metal-based materials, and carbon-based materials. The majority of clinically applicable drugs are liposomes, though other materials are currently undergoing continuous optimization. This review begins with the roles of autophagy in tumor treatment, and then focuses on the application of nanomaterials with autophagy-regulating functions in tumor treatment.
Collapse
Affiliation(s)
- Ketai He
- Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- West China School of Stomatology, Sichuan University, Sichuan, China
| | - Mingkun Chen
- West China School of Stomatology, Sichuan University, Sichuan, China
| | - Jiao Liu
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Sichuan, China
| | - Shufang Du
- West China School of Stomatology, Sichuan University, Sichuan, China
| | - Changyu Ren
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Sichuan, China
| | - Jifa Zhang
- Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
15
|
Liao Z, Liu X, Fan D, Sun X, Zhang Z, Wu P. Autophagy-mediated nanomaterials for tumor therapy. Front Oncol 2023; 13:1194524. [PMID: 38192627 PMCID: PMC10773885 DOI: 10.3389/fonc.2023.1194524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/30/2023] [Indexed: 01/10/2024] Open
Abstract
Autophagy is a lysosomal self-degradation pathway that plays an important protective role in maintaining intracellular environment. Deregulation of autophagy is related to several diseases, including cancer, infection, neurodegeneration, aging, and heart disease. In this review, we will summarize recent advances in autophagy-mediated nanomaterials for tumor therapy. Firstly, the autophagy signaling pathway for tumor therapy will be reviewed, including oxidative stress, mammalian target of rapamycin (mTOR) signaling and autophagy-associated genes pathway. Based on that, many autophagy-mediated nanomaterials have been developed and applied in tumor therapy. According to the different structure of nanomaterials, we will review and evaluate these autophagy-mediated nanomaterials' therapeutic efficacy and potential clinical application.
Collapse
Affiliation(s)
- Zijian Liao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
| | - Dianfa Fan
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
| | - Xingjun Sun
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
| | - Zhikun Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
| | - Pan Wu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
16
|
Sadeghi MS, Sangrizeh FH, Jahani N, Abedin MS, Chaleshgari S, Ardakan AK, Baeelashaki R, Ranjbarpazuki G, Rahmanian P, Zandieh MA, Nabavi N, Aref AR, Salimimoghadam S, Rashidi M, Rezaee A, Hushmandi K. Graphene oxide nanoarchitectures in cancer therapy: Drug and gene delivery, phototherapy, immunotherapy, and vaccine development. ENVIRONMENTAL RESEARCH 2023; 237:117027. [PMID: 37659647 DOI: 10.1016/j.envres.2023.117027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/19/2023] [Accepted: 08/29/2023] [Indexed: 09/04/2023]
Abstract
The latest advancements in oncology involves the creation of multifunctional nanostructures. The integration of nanoparticles into the realm of cancer therapy has brought about a transformative shift, revolutionizing the approach to addressing existing challenges and limitations in tumor elimination. This is particularly crucial in combating the emergence of resistance, which has significantly undermined the effectiveness of treatments like chemotherapy and radiotherapy. GO stands as a carbon-derived nanoparticle that is increasingly finding utility across diverse domains, notably in the realm of biomedicine. The utilization of GO nanostructures holds promise in the arena of oncology, enabling precise transportation of drugs and genetic material to targeted sites. GO nanomaterials offer the opportunity to enhance the pharmacokinetic behavior and bioavailability of drugs, with documented instances of these nanocarriers elevating drug accumulation at the tumor location. The GO nanostructures encapsulate genes, shielding them from degradation and facilitating their uptake within cancer cells, thereby promoting efficient gene silencing. The capability of GO to facilitate phototherapy has led to notable advancements in reducing tumor progression. By PDT and PTT combination, GO nanomaterials hold the capacity to diminish tumorigenesis. GO nanomaterials have the potential to trigger both cellular and innate immunity, making them promising contenders for vaccine development. Additionally, types of GO nanoparticles that respond to specific stimuli have been applied in cancer eradication, as well as for the purpose of cancer detection and biomarker diagnosis. Endocytosis serves as the mechanism through which GO nanomaterials are internalized. Given these advantages, the utilization of GO nanomaterials for tumor elimination comes highly recommended.
Collapse
Affiliation(s)
- Mohammad Saleh Sadeghi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Negar Jahani
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mahdi Sadegh Abedin
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Soheila Chaleshgari
- Department of Avian Diseases, Faculty of Veterinary Medicine, Chamran University, Ahvaz, Iran
| | - Alireza Khodaei Ardakan
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Reza Baeelashaki
- Department of Food Hygiene and Quality Control, Division of Animal Feed Hygiene, Faculty of Veterinary Medicine, Islamic Azad University, Shabestar Branch, Shabestar, Iran
| | - Golnaz Ranjbarpazuki
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parham Rahmanian
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Noushin Nabavi
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Amir Reza Aref
- Department of Cancer Biology, Center for Cancer Systems Biology, Dana-Farber Cancer Institute, Department of Genetics, Harvard Medical School, Boston, MA, USA; Department of Translational Sciences, Xsphera Biosciences Inc. Boston, MA, USA
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
17
|
Cerverò-Varona A, Canciello A, Peserico A, Haidar Montes AA, Citeroni MR, Mauro A, Russo V, Moffa S, Pilato S, Di Giacomo S, Dufrusine B, Dainese E, Fontana A, Barboni B. Graphene oxide accelerates TGFβ-mediated epithelial-mesenchymal transition and stimulates pro-inflammatory immune response in amniotic epithelial cells. Mater Today Bio 2023; 22:100758. [PMID: 37600353 PMCID: PMC10432246 DOI: 10.1016/j.mtbio.2023.100758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/23/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023] Open
Abstract
The application of biomaterials on immune regenerative strategies to deal with unsolved pathologies is getting attention in the field of tissue engineering. In this context, graphene oxide (GO) has been proposed as an immune-mimetic material largely used for developing stem cell-based regenerative therapies, since it has shown to influence stem cell behavior and modulate their immune response. Similarly, amniotic epithelial stem cells (AECs) are getting an increasing clinical interest as source of stem cells due to their great plasticity and immunomodulatory paracrine activities, even though GO bio-mimetic effects still remain unknown. To this aim, GO-functionalized glass coverslips have been used for AECs culture. The results demonstrated how GO-coating is able to induce and accelerate the Epithelial-Mesenchymal Transition (EMT), in a process mediated by the intracellular activation of TGFβ1-SMAD2/3 signaling pathway. The trans-differentiation towards mesenchymal phenotype provides AECs of migratory ability and substantially changes the pattern of cytokines secretion upon inflammatory stimulus. Indeed, GO-exposed AECs enhance their pro-inflammatory interleukins production thus inducing a more efficient activation of macrophages and, at the same time, by slightly reducing their inhibitory action on peripheral blood mononuclear cells proliferation. Therefore, the adhesion of AECs on GO-functionalized surfaces might contribute to the generation of a tailored microenvironment useful to face both the phases of the inflammation, thereby fostering the regenerative process.
Collapse
Affiliation(s)
- Adrian Cerverò-Varona
- Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo,64100, Teramo, Italy
| | - Angelo Canciello
- Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo,64100, Teramo, Italy
| | - Alessia Peserico
- Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo,64100, Teramo, Italy
| | - Arlette Alina Haidar Montes
- Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo,64100, Teramo, Italy
| | - Maria Rita Citeroni
- Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo,64100, Teramo, Italy
| | - Annunziata Mauro
- Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo,64100, Teramo, Italy
| | - Valentina Russo
- Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo,64100, Teramo, Italy
| | - Samanta Moffa
- Department of Pharmacy, University “G. D’Annunzio”, Via Dei Vestini, 66100, Chieti, Italy
| | - Serena Pilato
- Department of Pharmacy, University “G. D’Annunzio”, Via Dei Vestini, 66100, Chieti, Italy
| | - Stefano Di Giacomo
- Department of Pharmacy, University “G. D’Annunzio”, Via Dei Vestini, 66100, Chieti, Italy
| | - Beatrice Dufrusine
- Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo,64100, Teramo, Italy
| | - Enrico Dainese
- Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo,64100, Teramo, Italy
| | - Antonella Fontana
- Department of Pharmacy, University “G. D’Annunzio”, Via Dei Vestini, 66100, Chieti, Italy
| | - Barbara Barboni
- Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo,64100, Teramo, Italy
| |
Collapse
|
18
|
Zeng XY, Qiu XZ, Wu JN, Liang SM, Huang JA, Liu SQ. Interaction mechanisms between autophagy and ferroptosis: Potential role in colorectal cancer. World J Gastrointest Oncol 2023; 15:1135-1148. [PMID: 37546557 PMCID: PMC10401467 DOI: 10.4251/wjgo.v15.i7.1135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/28/2023] [Accepted: 04/23/2023] [Indexed: 07/12/2023] Open
Abstract
Colorectal cancer (CRC) is a common malignancy that has the second highest incidence and mortality rate. Although there are many personalized treatment options for CRC, the therapeutic effects are ultimately limited by drug resistance. Studies have aimed to block the initiation and progression of CRC by inducing cell death to overcome this obstacle. Substantial evidence has indicated that both autophagy and ferroptosis play important regulatory roles in CRC. Autophagy, a lysosome-dependent process by which cellular proteins and organelles are degraded, is the basic mechanism for maintaining cell homeostasis. The duality and complexity of autophagy in cancer therapy is a hot topic of discussion. Ferroptosis, a regulated cell death pathway, is associated with iron accumulation-induced lipid peroxidation. The activation of ferroptosis can suppress CRC proliferation, invasion and drug resistance. Furthermore, recent studies have suggested an interaction between autophagy and ferroptosis. Autophagy can selectively degrade certain cellular contents to provide raw materials for ferroptosis, ultimately achieving antitumor and anti-drug resistance. Therefore, exploring the interaction between autophagy and ferroptosis could reveal novel ideas for the treatment of CRC. In this review, we describe the mechanisms of autophagy and ferroptosis, focusing on their roles in CRC and the crosstalk between them.
Collapse
Affiliation(s)
- Xin-Ya Zeng
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Xin-Ze Qiu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Jiang-Ni Wu
- Department of Pathology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Sheng-Mei Liang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Jie-An Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Shi-Quan Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
19
|
Daniluk K, Lange A, Wójcik B, Zawadzka K, Bałaban J, Kutwin M, Jaworski S. Effect of Melittin Complexes with Graphene and Graphene Oxide on Triple-Negative Breast Cancer Tumors Grown on Chicken Embryo Chorioallantoic Membrane. Int J Mol Sci 2023; 24:ijms24098388. [PMID: 37176095 PMCID: PMC10179033 DOI: 10.3390/ijms24098388] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
One of the components of bee venom is melittin (M), which has strong lysing properties on membranes. M has high toxicity to cancer cells, but it also affects healthy cells, making it necessary to use methods for targeted delivery to ensure treatment. This research is a continuation of previous studies using graphene nanomaterials as M carriers to breast cancer cells. The studies described below are conducted on a more organized biological structure than what is found in vitro cells, namely, cancerous tumors grown on a chicken embryo chorioallantoic membrane. Caspase 3 and 8 levels are analyzed, and the level of oxidative stress markers and changes in protein expression for cytokines are examined. The results show that M complexes with nanomaterials reduce the level of oxidative stress more than M alone does, but the use of graphene (GN) as a carrier increases the level of DNA damage to a greater extent than the increase caused by M alone. An analysis of cytokine levels shows that the use of the M and GN complex increases the level of proteins responsible for inhibiting tumor progression to a greater extent than the increase occasioned by a complex with graphene oxide (GO). The results suggest that the use of GN as an M carrier may increase the toxic effect of M on structures located inside a cell.
Collapse
Affiliation(s)
- Karolina Daniluk
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Agata Lange
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Barbara Wójcik
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Katarzyna Zawadzka
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Jaśmina Bałaban
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Marta Kutwin
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Sławomir Jaworski
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| |
Collapse
|
20
|
Taheriazam A, Abad GGY, Hajimazdarany S, Imani MH, Ziaolhagh S, Zandieh MA, Bayanzadeh SD, Mirzaei S, Hamblin MR, Entezari M, Aref AR, Zarrabi A, Ertas YN, Ren J, Rajabi R, Paskeh MDA, Hashemi M, Hushmandi K. Graphene oxide nanoarchitectures in cancer biology: Nano-modulators of autophagy and apoptosis. J Control Release 2023; 354:503-522. [PMID: 36641122 DOI: 10.1016/j.jconrel.2023.01.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 01/16/2023]
Abstract
Nanotechnology is a growing field, with many potential biomedical applications of nanomedicine for the treatment of different diseases, particularly cancer, on the horizon. Graphene oxide (GO) nanoparticles can act as carbon-based nanocarriers with advantages such as a large surface area, good mechanical strength, and the capacity for surface modification. These nanostructures have been extensively used in cancer therapy for drug and gene delivery, photothermal therapy, overcoming chemotherapy resistance, and for imaging procedures. In the current review, we focus on the biological functions of GO nanoparticles as regulators of apoptosis and autophagy, the two major forms of programmed cell death. GO nanoparticles can either induce or inhibit autophagy in cancer cells, depending on the conditions. By stimulating autophagy, GO nanocarriers can promote the sensitivity of cancer cells to chemotherapy. However, by impairing autophagy flux, GO nanoparticles can reduce cell survival and enhance inflammation. Similarly, GO nanomaterials can increase ROS production and induce DNA damage, thereby sensitizing cancer cells to apoptosis. In vitro and in vivo experiments have investigated whether GO nanomaterials show any toxicity in major body organs, such as the brain, liver, spleen, and heart. Molecular pathways, such as ATG, MAPK, JNK, and Akt, can be regulated by GO nanomaterials, leading to effects on autophagy and apoptosis. These topics are discussed in this review to shed some lights towards the biomedical potential of GO nanoparticles and their biocompatibility, paving the way for their future application in clinical trials.
Collapse
Affiliation(s)
- Afshin Taheriazam
- Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Ghazaleh Gholamiyan Yousef Abad
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Shima Hajimazdarany
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Hassan Imani
- Department of Clinical Science, Faculty of Veterinary Medicine, Islamic Azad University, Shahr-e kord Branch, Chaharmahal and Bakhtiari, Iran
| | - Setayesh Ziaolhagh
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa; Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Vice President at Translational Sciences, Xsphera Biosciences Inc., 6 Tide Street, Boston, MA, 02210, USA
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkey
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Romina Rajabi
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Mahshid Deldar Abad Paskeh
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|