1
|
Li L, Liu B, Zhang H, Wang C, Sun L, Zhang Y, Song L, Yu Y, Zhou K. 4-Phenylbutyric acid suppresses psoralen-induced hepatotoxicity by inhibiting ERS and reestablishing mitochondrial fusion-fission balance in mice. Toxicology 2024; 509:153954. [PMID: 39299507 DOI: 10.1016/j.tox.2024.153954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/06/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
Psoralen is a main active molecule of the traditional Chinese herb medicine Fructus Psoraleae. Our previous studies have shown that psoralen induced liver injury through the endoplasmic reticulum stress (ERS) signaling pathways. In this article, we studied whether the ERS inhibitor, 4-phenylbutyrate acid (4-PBA) could inhibit the liver toxicity caused by psoralen, and explored the underlying mechanisms. Mice were given the solvent, 20 mg/kg, 40 mg/kg, 80 mg/kg of psoralen, or 80 mg/kg of psoralen plus 4-PBA for 14 days. We found that 4-PBA significantly reduced the serum LDH and liver tissue MDA level, increased the activities of SOD and CAT, reduced liver weight and coefficient, repaired histopathological damage, and inhibited hepatocytes apoptosis induced by psoralen. RNA-seq transcriptomics found that except for the endoplasmic reticulum, the mitochondria was severely affected by psoralen. And genes involved in mitochondrial fusion, apoptosis, protein folding, and autophagy were found differently expressed in the psoralen group. Further studies found that 4-PBA inhibited the overexpression of GRP78 and CHOP, increased the Bcl-2/Bax ratio, and reduced the expression of Caspase-3. Moreover, 4-PBA reduced the overexpression of mitochondrial fission protein DRP1, increased the expression of fusion proteins Mfn-2 and OPA1, but has no inhibitory effects on autophagy proteins Atg5 or LC3A/B. In conclusion, 4-PBA inhibited ERS and reestablished mitochondrial fusion-fission balance, thereby blocking cell apoptosis, oxidative stress, and mitochondrial dysfunction, thus prevented against psoralen-induced hepatotoxicity.
Collapse
Affiliation(s)
- Li Li
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Bing Liu
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Haorui Zhang
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chen Wang
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Likang Sun
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yue Zhang
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin 301617, China
| | - Lei Song
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin 301617, China
| | - Yingli Yu
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin 301617, China.
| | - Kun Zhou
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
2
|
Iliadis S, Papanikolaou NA. Reactive Oxygen Species Mechanisms that Regulate Protein-Protein Interactions in Cancer. Int J Mol Sci 2024; 25:9255. [PMID: 39273204 PMCID: PMC11395503 DOI: 10.3390/ijms25179255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Reactive oxygen species (ROS) are produced during cellular metabolism and in response to environmental stress. While low levels of ROS play essential physiological roles, excess ROS can damage cellular components, leading to cell death or transformation. ROS can also regulate protein interactions in cancer cells, thereby affecting processes such as cell growth, migration, and angiogenesis. Dysregulated interactions occur via various mechanisms, including amino acid modifications, conformational changes, and alterations in complex stability. Understanding ROS-mediated changes in protein interactions is crucial for targeted cancer therapies. In this review, we examine the role that ROS mechanisms in regulating pathways through protein-protein interactions.
Collapse
Affiliation(s)
- Stavros Iliadis
- Laboratory of Biological Chemistry, Department of Medicine, Section of Biological Sciences and Preventive Medicine, Aristotle University of Thessaloniki School of Medicine, 54124 Thessaloniki, Macedonia, Greece
| | - Nikolaos A Papanikolaou
- Laboratory of Biological Chemistry, Department of Medicine, Section of Biological Sciences and Preventive Medicine, Aristotle University of Thessaloniki School of Medicine, 54124 Thessaloniki, Macedonia, Greece
| |
Collapse
|
3
|
Zoughaib M, Pashirova TN, Nikolaeva V, Kamalov M, Nakhmetova F, Salakhieva DV, Abdullin TI. Anticancer and Chemosensitizing Effects of Menadione-Containing Peptide-Targeted Solid Lipid Nanoparticles. J Pharm Sci 2024; 113:2258-2267. [PMID: 38508340 DOI: 10.1016/j.xphs.2024.03.009] [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: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
Vitamin K derivatives such as menadione (MD) have been recognized as promising redox-modulating and chemosensitizing agents for anticancer therapy, however, their cellular activities in peptide-targeted nanocarriers have not been elucidated to date. This study provides the guidelines for developing MD-loaded solid lipid nanoparticles (SLN) modified with extracellular matrix (ECM)-derived peptides. Relationships between RGD peptide concentration and changes in DLS characteristics as well as accumulation of SLN in cancer cells were revealed to adjust the peptide-lipid ratio. SLN system maintained adequate nanoparticle concentration and low dispersity after introduction of MD and MD/RGD, whereas formulated MD was protected from immediate conjugation with reduced glutathione (GSH). RGD-modified MD-containing SLN showed enhanced prooxidant, GSH-depleting and cytotoxic activities toward PC-3 prostate cancer cells attributed to improved cellular pharmacokinetics of the targeted formulation. Furthermore, this formulation effectively sensitized PC-3 cells and OVCAR-4 ovarian cancer cells to free doxorubicin and cisplatin so that cell growth was inhibited by MD-drug composition at nontoxic concentrations of the ingredients. These results provide an important background for further improving chemotherapeutic methods based on combination of conventional cytostatics with peptide-targeted SLN formulations of MD.
Collapse
Affiliation(s)
- Mohamed Zoughaib
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia; Scientific and Educational Center of Pharmaceutics, Kazan Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia.
| | - Tatiana N Pashirova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov St., 420088 Kazan, Russia
| | - Viktoriia Nikolaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia; Scientific and Educational Center of Pharmaceutics, Kazan Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Marat Kamalov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia; Scientific and Educational Center of Pharmaceutics, Kazan Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Fidan Nakhmetova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia; Scientific and Educational Center of Pharmaceutics, Kazan Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Diana V Salakhieva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia; Scientific and Educational Center of Pharmaceutics, Kazan Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Timur I Abdullin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia; Scientific and Educational Center of Pharmaceutics, Kazan Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia.
| |
Collapse
|
4
|
Inacio KK, Pessoa ADS, Tokuhara CK, Pagnan AL, Sanches MLR, Fakhoury VS, Oliveira GSND, Oliveira FAD, Ximenes VF, Oliveira RCD. Menadione and protocatechuic acid: A drug combination with antitumor effects in murine osteosarcoma cells. Arch Biochem Biophys 2024; 751:109840. [PMID: 38040223 DOI: 10.1016/j.abb.2023.109840] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/22/2023] [Accepted: 11/25/2023] [Indexed: 12/03/2023]
Abstract
Osteosarcoma (OS) is a primary malignant bone tumor that has an abnormal expression of oncogenesis and tumor suppressors and causes dysregulation of various signaling pathways. Thus, novel therapeutic strategies for OS are needed to overcome the resistance of traditional treatments. This study evaluated the cytotoxic and anticancer effects of the association between menadione (MEN) and protocatechuic acid (PCA) in murine OS cells (UMR-106). The concentrations were 3.12 μM of isolated MEN, 500 μM of isolated PCA, and their associations. We performed cell viability assays, morphology modification analysis, cell migration by the wound-healing method, apoptosis by flow cytometry, reactive oxygen species (ROS) production, gene expression of NOX by RT-qPCR, and degradation of MMP-2 and 9 by zymography. Our results showed that the association of MEN+PCA was more effective in OS cells than the compounds alone. The association decreased cell viability, delayed cell migration, and decreased the expression of NOX-2 and ROS. In addition, the MEN+PCA association induced a slight increase in the apoptotic process. In summary, the association can enhance the compound's antitumor effects and establish a higher selectivity for tumor cells, possibly caused by significant mitochondrial damage and antioxidant properties.
Collapse
Affiliation(s)
- Kelly Karina Inacio
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Brazil
| | - Adriano de Souza Pessoa
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Brazil
| | - Cintia Kazuko Tokuhara
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Brazil
| | - Ana Lígia Pagnan
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Brazil
| | | | | | | | - Flavia Amadeu de Oliveira
- Sanford Children's Health Research Center. Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Valdecir Farias Ximenes
- Department of Chemistry, Faculty of Sciences, UNESP, São Paulo State University, Bauru, São Paulo, Brazil
| | | |
Collapse
|
5
|
Sheikh E, Agrawal K, Roy S, Burk D, Donnarumma F, Ko YH, Guttula PK, Biswal NC, Shukla HD, Gartia MR. Multimodal Imaging of Pancreatic Cancer Microenvironment in Response to an Antiglycolytic Drug. Adv Healthc Mater 2023; 12:e2301815. [PMID: 37706285 PMCID: PMC10842640 DOI: 10.1002/adhm.202301815] [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: 07/03/2023] [Indexed: 09/15/2023]
Abstract
Lipid metabolism and glycolysis play crucial roles in the progression and metastasis of cancer, and the use of 3-bromopyruvate (3-BP) as an antiglycolytic agent has shown promise in killing pancreatic cancer cells. However, developing an effective strategy to avoid chemoresistance requires the ability to probe the interaction of cancer drugs with complex tumor-associated microenvironments (TAMs). Unfortunately, no robust and multiplexed molecular imaging technology is currently available to analyze TAMs. In this study, the simultaneous profiling of three protein biomarkers using SERS nanotags and antibody-functionalized nanoparticles in a syngeneic mouse model of pancreatic cancer (PC) is demonstrated. This allows for comprehensive information about biomarkers and TAM alterations before and after treatment. These multimodal imaging techniques include surface-enhanced Raman spectroscopy (SERS), immunohistochemistry (IHC), polarized light microscopy, second harmonic generation (SHG) microscopy, fluorescence lifetime imaging microscopy (FLIM), and untargeted liquid chromatography and mass spectrometry (LC-MS) analysis. The study reveals the efficacy of 3-BP in treating pancreatic cancer and identifies drug treatment-induced lipid species remodeling and associated pathways through bioinformatics analysis.
Collapse
Affiliation(s)
- Elnaz Sheikh
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Kirti Agrawal
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Sanjit Roy
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - David Burk
- Department of Cell Biology and Bioimaging, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Fabrizio Donnarumma
- Department of Chemistry, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Young H Ko
- NewG Lab Pharma, 701 East Pratt Street, Columbus Center, Baltimore, MD, 21202, USA
| | - Praveen Kumar Guttula
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Nrusingh C Biswal
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Hem D Shukla
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Manas Ranjan Gartia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA, 70803, USA
| |
Collapse
|
6
|
Davidescu M, Mezzasoma L, Fettucciari K, Pascucci L, Pariano M, Di Michele A, Bereshchenko O, Cagini C, Cellini B, Corazzi L, Bellezza I, Macchioni L. Cardiolipin-mediated temporal response to hydroquinone toxicity in human retinal pigmented epithelial cell line. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119554. [PMID: 37524263 DOI: 10.1016/j.bbamcr.2023.119554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 08/02/2023]
Abstract
Hydroquinone, a potent toxic agent of cigarette smoke, damages retinal pigmented epithelial cells by triggering oxidative stress and mitochondrial dysfunction, two events causally related to the development and progression of retinal diseases. The inner mitochondrial membrane is enriched in cardiolipin, a phospholipid susceptible of oxidative modifications which determine cell-fate decision. Using ARPE-19 cell line as a model of retinal pigmented epithelium, we analyzed the potential involvement of cardiolipin in hydroquinone toxicity. Hydroquinone exposure caused an early concentration-dependent increase in mitochondrial reactive oxygen species, decrease in mitochondrial membrane potential, and rise in the rate of oxygen consumption not accompanied by changes in ATP levels. Despite mitochondrial impairment, cell viability was preserved. Hydroquinone induced cardiolipin translocation to the outer mitochondrial membrane, and an increase in the colocalization of the autophagosome adapter protein LC3 with mitochondria, indicating the induction of protective mitophagy. A prolonged hydroquinone treatment induced pyroptotic cell death by cardiolipin-mediated caspase-1 and gasdermin-D activation. Cardiolipin-specific antioxidants counteracted hydroquinone effects pointing out that cardiolipin can act as a mitochondrial "eat-me signal" or as a pyroptotic cell death trigger. Our results indicate that cardiolipin may act as a timer for the mitophagy to pyroptosis switch and propose cardiolipin-targeting compounds as promising approaches for the treatment of oxidative stress-related retinal diseases.
Collapse
Affiliation(s)
- Magdalena Davidescu
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Letizia Mezzasoma
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Katia Fettucciari
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, Via S. Costanzo 4, 06126 Perugia, Italy
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Alessandro Di Michele
- Department of Physic and Geology, University of Perugia, Via Pascoli, Perugia 06123, Italy
| | - Oxana Bereshchenko
- Department of Philosophy, Social Sciences, Humanities and Education, University of Perugia, Piazza Ermini 1, Perugia 06123, Italy
| | - Carlo Cagini
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Barbara Cellini
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Lanfranco Corazzi
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Ilaria Bellezza
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Lara Macchioni
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy.
| |
Collapse
|
7
|
Gowda BHJ, Ahmed MG, Alshehri SA, Wahab S, Vora LK, Singh Thakur RR, Kesharwani P. The cubosome-based nanoplatforms in cancer therapy: Seeking new paradigms for cancer theranostics. ENVIRONMENTAL RESEARCH 2023; 237:116894. [PMID: 37586450 DOI: 10.1016/j.envres.2023.116894] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/10/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
Lyotropic liquid crystals are self-assembled, non-lamellar, and mesophase nanostructured materials that have garnered significant attention as drug carriers. Cubosomes, a subtype of lyotropic liquid crystalline nanoparticles, possess three-dimensional structures that display bicontinuous cubic liquid-crystalline patterns. These patterns are formed through the self-organization of unsaturated monoglycerides (amphphilic lipids such as glyceryl monooleate or phytantriol), followed by stabilization using steric polymers (poloxamers). Owing to their bicontinuous structure and steric polymer-based stabilization, cubosomes have been demonstrated to possess greater entrapment efficiency for hydrophobic drugs compared to liposomes, while also exhibiting high stability. In the past decade, there has been significant interest in cubosomes due to their ability to deliver therapeutic and contrast agents for cancer treatment and imaging with minimal side effects, establishing them as a safe and effective approach. Concerning these advantages, the present review elaborates on the general aspects, composition, and preparation techniques of cubosomes, followed by explanations of their mechanisms of drug loading and release patterns. Furthermore, the review provides meticulous discussions on the use of cubosomes in the treatment and imaging of various types of cancer, culminating in the enumeration of patents related to cubosome-based drug delivery systems.
Collapse
Affiliation(s)
- B H Jaswanth Gowda
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, Karnataka, India
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, Karnataka, India
| | - Saad Ali Alshehri
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, United Kingdom
| | - Raghu Raj Singh Thakur
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, United Kingdom
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Center for Global health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| |
Collapse
|
8
|
He Y, Chen H, Li W, Xu L, Yao H, Cao Y, Wang Z, Zhang L, Wang D, Zhou D. 3-Bromopyruvate-loaded bismuth sulfide nanospheres improve cancer treatment by synergizing radiotherapy with modulation of tumor metabolism. J Nanobiotechnology 2023; 21:209. [PMID: 37408010 DOI: 10.1186/s12951-023-01970-8] [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: 05/11/2023] [Accepted: 06/28/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND Radiotherapy (RT) is one of the most mainstream cancer therapeutic modalities. However, due to the lack of specificity of the radiation adopted, both normal and cancerous cells are destroyed indiscriminately. This highlights the crucial need to improve radiosensitization. This study aims to address this issue by constructing a multifunctional nanospheres that can sensitize multiple aspects of radiotherapy. RESULTS Nanospheres containing high atomic element Bi can effectively absorb ionizing radiation and can be used as radiosensitizers. Cell viability after Bi2S3 + X-ray treatment was half that of X-ray treatment alone. On the other hand, exposed 3-bromopyruvate (3BP) could reduce the overactive oxygen (O2) metabolism of tumor cells and alleviate tumor hypoxia, thereby promoting radiation-induced DNA damage. The combination index (CI) of 3BP and Bi2S3-based RT in Bi2S3-3BP + X-ray was determined to be 0.46 with the fraction affected (fa) was 0.5 via Chou-Talalay's isobolographic method, which indicated synergistic effect of 3BP and Bi2S3-based RT after integration into Bi2S3-3BP + X-ray. Under the combined effect of 3BP and RT, autophagy was over-activated through starvation-induced and redox homeostasis dysregulation pathways, which in turn exhibited pro-death effects. In addition, the prepared nanospheres possess strong X-ray attenuation and high near-infrared (NIR) optical absorption, thus eliminating the need for additional functional components and could serve as bimodal contrast agents for computed tomography/photoacoustic (CT/PA) imaging. CONCLUSIONS The rational design of multifunctional nanospheres with the unique properties provided a novel strategy to achieving high therapeutic efficacy in RT. This was accomplished through simultaneous activation of multiple sensitization pathways by increasing ionizing radiation, reducing tumor oxygen consumption, inducing pro-death autophagy, and providing multiple-imaging guidance/monitoring.
Collapse
Affiliation(s)
- Yiman He
- Department of Ultrasound, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, P.R. China
| | - Huawan Chen
- Department of Oncology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, P.R. China
| | - Wenbo Li
- Department of Nuclear Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, P.R. China
| | - Lu Xu
- Department of Nuclear Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, P.R. China
| | - Huan Yao
- Department of Ultrasound, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, P.R. China
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, P.R. China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, P.R. China
| | - Liang Zhang
- Department of Ultrasound, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, P.R. China
| | - Dong Wang
- Department of Ultrasound, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, P.R. China.
| | - Di Zhou
- Department of Radiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, P.R. China.
| |
Collapse
|
9
|
Mireștean CC, Iancu RI, Iancu DPT. New horizons in modulating the radio-sensitivity of head and neck cancer - 100 years after Warburg' effect discovery. Front Oncol 2022; 12:908695. [PMID: 36568220 PMCID: PMC9780029 DOI: 10.3389/fonc.2022.908695] [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: 03/30/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Tumor radiation resistance along with chemotherapy resistance is one of the main causes of therapeutic failure of radiotherapy-treated head and neck cancers. 100 years after the discovery of the Warburg effect, a process specific to malignant cells to metabolize glucose especially anaerobically even under normoxia condition, its modulation has become a viable therapeutic target for improving the results of cancer therapies. Improving the radio-sensitivity of head and neck tumors by reversing the Warburg effect can increase the rate of local control and reduce the toxicity associated with irradiation. P53 status can be used as a biomarker in the choice of a single agent strategy (cell respiration inhibition with Metformin) or double inhibition, both of respiration and glycolysis. Targeting of enzymes involved in the Warburg effect, such as Hexokinase-II, are strategies with potential to be applied in clinical practice with radio-sensitizing effect for head and neck squamous cell carcinoma. Even if anti-Warburg therapies tested in clinical trials have been associated with either toxic deaths or a minor clinical benefit, the identification of both potential radio-sensitivity biomarkers and methods of reversing the Warburg effect will play an important role in the radiobiology of head and neck cancers.
Collapse
Affiliation(s)
- Camil Ciprian Mireștean
- Department of Medical Oncology and Radiotherapy, University of Medicine and Pharmacy Craiova, Craiova, Romania,Department of Surgery, Railways Clinical Hospital, Iasi, Romania
| | - Roxana Irina Iancu
- Oral Pathology Department, “Gr.T.Popa” University of Medicine and Pharmacy, Iasi, Romania,Department of Clinical Laboratory, St. Spiridon Emergency Hospital, Iasi, Romania,*Correspondence: Roxana Irina Iancu,
| | - Dragoș Petru Teodor Iancu
- Department of Medical Oncology and Radiotherapy, “Gr.T.Popa” University of Medicine and Pharmacy, Iasi, Romania,Department of Radiation Oncology, Regional Institute of Oncology, Iasi, Romania
| |
Collapse
|
10
|
3-Bromopyruvate inhibits pancreatic tumor growth by stalling glycolysis, and dismantling mitochondria in a syngeneic mouse model. Am J Cancer Res 2022; 12:4977-4987. [PMID: 36504891 PMCID: PMC9729896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/25/2022] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer (PC) is the fourth-most-deadly cancer in the United States with a 5-year survival rate of only 8%. The majority of patients with locally advanced pancreatic cancer undergo chemotherapy and/or radiation therapy (RT). However, current treatments are inadequate and novel strategies are desperately required. 3-Bromopyruvate (3-BP) is a promising anticancer drug against pancreatic cancer. It exerts potent anticancer effects by inhibiting hexokinase II enzyme (HK2) of the glycolytic pathway in cancer cells while not affecting the normal cells. 3-BP killed 95% of Panc-2 cells at 15 μM concentration and severely inhibited ATP production by disrupting the interaction between HK2 and mitochondrial Voltage Dependent Anion Channel-1 (VDAC1) protein. Electron microscopy data revealed that 3-BP severely damaged mitochondrial membrane in cancer cells. We further examined therapeutic effect of 3-BP in syngeneic mouse pancreatic cancer model by treating animals with 10, 15 and 20 mg/kg dose. 3-BP at 15 & 20 mg/kg dose level significantly reduced tumor growth by approximately 75-80% in C57BL/6 female mice. Immunohistochemistry data showed complete inhibition of hexokinase II (HK2) and TGFβ, in animals treated with 3-BP drug. We also observed enhanced expression of active caspase-3 in tumor tissues exhibited apoptotic death. Flow Cytometry analysis showed significant inhibition in MDSC (CD11b) population in treated tumor which may have allowed infiltration of CD8+ T cells and inhibited tumor growth. Notably, metabolomic data also revealed severe inhibition in glycolysis, NADP, ATP and lactic acid production in cancer cells treated with 40 μM 3-BP. Importantly, we also observed inhibition in lactic acid production responsible for tumor aggression. These results provide new evidence that 3-BP severely inhibit glucose metabolism in cancer cells by blocking hexokinase II, and disrupting mitochondria by suppressing BCL2L1 in pancreatic cancer.
Collapse
|
11
|
Plumeria alba-Mediated Green Synthesis of Silver Nanoparticles Exhibits Antimicrobial Effect and Anti-Oncogenic Activity against Glioblastoma U118 MG Cancer Cell Line. NANOMATERIALS 2022; 12:nano12030493. [PMID: 35159838 PMCID: PMC8839720 DOI: 10.3390/nano12030493] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/23/2022] [Accepted: 01/28/2022] [Indexed: 12/19/2022]
Abstract
Plumeria alba (P. alba) is a small laticiferous tree with promising medicinal properties. Green synthesis of nanoparticles is eco-friendly, cost-effective, and non-hazardous compared to chemical and physical synthesis methods. Current research aiming to synthesize silver nanoparticles (AgNPs) from the leaf extract of P. alba (P- AgNPs) has described its physiochemical and pharmacological properties in recognition of its therapeutic potential as an anticancer and antimicrobial agent. These biogenic synthesized P-AgNPs were physiochemically characterized by ultraviolet-visible spectroscopy, Fourier-transform infrared spectroscopy (FTIR), scanning electron microscopy (SEM), transmission electron microscope (TEM), atomic force microscopy (AFM), X-ray diffractometry (XRD), and zeta potential analyses. Antimicrobial activity was investigated against Escherichia coli, Pseudomonas aeruginosa, Enterobacter aerogenes, Enterococcus faecalis, Bacillus subtilis, Streptococcus pneumoniae, Candida albicans, and Candida glabrata. Anticancer activity against glioblasoma U118 MG cancer lines was investigated using an MTT assay, and apoptosis activity was determined by flow cytometry. UV–visible spectroscopic analysis portrayed surface plasmon resonance at 403 nm of synthesized P-AgNPs, and FTIR suggested the presence of amines, alkanes, and phenol molecules that could be involved in reduction and capping processes during AgNPs formation. Synthesized particles were spherical in shape and poly-dispersed with an average particle size of 26.43 nm and a poly-dispersity index (PDI) of 0.25 with a zeta potential value of −24.6 mV, ensuring their stability. The lattice plane values confirm the crystalline nature as identified by XRD. These P-AgNPs exhibited potential antimicrobial activity against selected human pathogenic microbes. Additionally, the in vitro MTT assay results show its effective anticancer activity against the glioma U118 MG cancer cell line with an IC50 value of 9.77 µg/mL AgNPs by initiating apoptosis as identified by a staining study with flow cytometric annexin V–fluorescein isothiocyanate (FITC) and propidium iodide (PI). Thus, P. alba AgNPs can be recommended for further pharmacological and other biological research. To conclude, the current investigation developed an eco-friendly AgNPs synthesis using P. alba leaf extract with potential cytotoxic and antibacterial capacity, which can therefore be recommended as a new strategy to treat different human diseases.
Collapse
|
12
|
Cattò C, Villa F, Cappitelli F. Understanding the Role of the Antioxidant Drug Erdosteine and Its Active Metabolite on Staphylococcus aureus Methicillin Resistant Biofilm Formation. Antioxidants (Basel) 2021; 10:antiox10121922. [PMID: 34943025 PMCID: PMC8698571 DOI: 10.3390/antiox10121922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 01/02/2023] Open
Abstract
Increasing numbers of researches have suggested that some drugs with reactive oxygen species (ROS)-mediated mechanisms of action modulate biofilm formation of some pathogenic strains. However, the full contribution of ROS to biofilm development is still an open question. In this paper, the correlations between the antioxidant drug Erdosteine (Er) and its active Metabolite I (Met I), ROS and biofilm development of two strains of methicillin resistant Staphylococcus aureus are presented. Experiments revealed that Er and Met I at 2 and 5 mg/L increased up to three orders of magnitude the number of biofilm-dwelling cells, while the content of ROS within the biofilms was reduced above the 87%, with a major effect of Met I in comparison to Er. Comparative proteomics showed that, 5 mg/L Met I modified the expression of 30% and 65% of total proteins in the two strains respectively. Some proteins involved in cell replication were upregulated, and a nitric oxide-based mechanism is assumed to modulate the biofilm development by changing quorum sensitive pathways. Additionally, several proteins involved in virulence were downregulated in the presence of Met I, suggesting that treated cells, despite being greater in number, might have lost part of their virulence.
Collapse
|
13
|
Yang X, Zheng Y, Liu L, Huang J, Wang F, Zhang J. Progress on the study of the anticancer effects of artesunate. Oncol Lett 2021; 22:750. [PMID: 34539854 PMCID: PMC8436334 DOI: 10.3892/ol.2021.13011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 07/30/2021] [Indexed: 12/16/2022] Open
Abstract
Artesunate (ART) is a derivative of artemisinin that is extracted from the wormwood plant Artemisia annua. ART is an antimalarial drug that has been shown to be safe and effective for clinical use. In addition to its antimalarial properties, ART has been attracting attention over recent years due to its reported inhibitory effects on cancer cell proliferation, invasion and migration. Therefore, ART has a wider range of potential clinical applications than first hypothesized. The aim of the present review was to summarize the latest research progress on the possible anticancer effects of ART, in order to lay a theoretical foundation for the further development of ART as a therapeutic option for cancer.
Collapse
Affiliation(s)
- Xiulan Yang
- Department of Pharmacology, The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Yudong Zheng
- Department of Pharmacology, The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Lian Liu
- Department of Pharmacology, The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Jiangrong Huang
- Department of Pharmacology, The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Fei Wang
- Center of Experiment and Training, Hubei College of Chinese Medicine, Jingzhou, Hubei 434020, P.R. China
| | - Jie Zhang
- Department of Pharmacology, The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| |
Collapse
|