1
|
Kaufman MJ, Meloni EG. Xenon gas as a potential treatment for opioid use disorder, alcohol use disorder, and related disorders. Med Gas Res 2025; 15:234-253. [PMID: 39812023 PMCID: PMC11918480 DOI: 10.4103/mgr.medgasres-d-24-00063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/26/2024] [Indexed: 01/16/2025] Open
Abstract
Xenon gas is considered to be a safe anesthetic and imaging agent. Research on its other potentially beneficial effects suggests that xenon may have broad efficacy for treating health disorders. A number of reviews on xenon applications have been published, but none have focused on substance use disorders. Accordingly, we review xenon effects and targets relevant to the treatment of substance use disorders, with a focus on opioid use disorder and alcohol use disorder. We report that xenon inhaled at subsedative concentrations inhibits conditioned memory reconsolidation and opioid withdrawal symptoms. We review work by others reporting on the antidepressant, anxiolytic, and analgesic properties of xenon, which could diminish negative affective states and pain. We discuss research supporting the possibility that xenon could prevent analgesic- or stress-induced opioid tolerance and, by so doing could reduce the risk of developing opioid use disorder. The rapid kinetics, favorable safety and side effect profiles, and multitargeting capability of xenon suggest that it could be used as an ambulatory on-demand treatment to rapidly attenuate maladaptive memory, physical and affective withdrawal symptoms, and pain drivers of substance use disorders when they occur. Xenon may also have human immunodeficiency virus and oncology applications because its effects relevant to substance use disorders could be exploited to target human immunodeficiency virus reservoirs, human immunodeficiency virus protein-induced abnormalities, and cancers. Although xenon is expensive, low concentrations exert beneficial effects, and gas separation, recovery, and recycling advancements will lower xenon costs, increasing the economic feasibility of its therapeutic use. More research is needed to better understand the remarkable repertoire of effects of xenon and its potential therapeutic applications.
Collapse
Affiliation(s)
- Marc J Kaufman
- McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | | |
Collapse
|
2
|
Lei Y, Dong R, Sun C, Hu Y, Yan Y, Song G, Wang Y. The Role of CXCL13 in GC-1 Cell Cycle Arrest Induced by Titanium Dioxide Nanoparticles Through JAK2/STAT3 Signaling Pathway. J Appl Toxicol 2025; 45:830-840. [PMID: 39777719 DOI: 10.1002/jat.4747] [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: 10/03/2024] [Revised: 12/02/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025]
Abstract
Titanium dioxide nanoparticles (TiO2 NPs) can induce the cell cycle arrest in spermatogonia, and the JAK2/STAT3 signaling pathway plays a pivotal role in cell cycle progression, but the specific upstream regulatory mechanisms are not completely clarified. The purpose of this study was to investigate whether CXCL13 regulated the JAK2/STAT3 signaling pathway to participate in cell cycle arrest after mouse spermatogonia cell line (GC-1) exposure to TiO2 NPs. The GC-1 cells were treated with TiO2 NPs at different concentrations (0, 10, 20, 30, and 40 μg/mL) for 24 h to detect cell viability, cell cycle distribution, CXCL13 protein, JAK2/STAT3 pathway-related proteins, and cell cycle-related proteins. The CXCL13 recombinant protein was used to verify the role of CXCL13 in cell cycle and JAK2/STAT3 signaling pathway. TiO2 NPs inhibited cell viability; regulated cell cycle-related proteins including remarkably decreased Cyclin D1, CDK4, Cyclin E1, and CDK2 as well as increased p21; and induced cell cycle arrest at the G0/G1 phase. TiO2 NPs inhibited the levels of CXCL13 protein and weakened the activation of the JAK2/STAT3 signaling pathway by reducing the levels of p-JAK2/JAK2 and p-STAT3/STAT3 proteins. Furthermore, CXCL13 mitigated the suppression of the JAK2/STAT3 signaling pathway and the G0/G1 cell cycle arrest caused by TiO2 NPs. Taken together, TiO2 NPs downregulated the expression of CXCL13 to inhibit the activation of downstream JAK2/STAT3 signaling pathway, eventually inducing cell cycle arrest at the G0/G1 phase. These results provide a novel insight for complemented understanding of the mechanisms of TiO2 NPs-induced cell cycle arrest in GC-1 cells.
Collapse
Affiliation(s)
- Yuzhu Lei
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Ruoyun Dong
- Xi'an Center for Disease Control and Prevention, Xi'an, China
| | - Chenhao Sun
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Yunhua Hu
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Yizhong Yan
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Guanling Song
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Yan Wang
- School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
3
|
Huang J, Liu F, Xu ZF, Xiang HL, Yuan Q, Zhang C. Minichromosome maintenance 4 plays a key role in protecting against acute kidney injury by regulating tubular epithelial cells survival and regeneration. J Adv Res 2025:S2090-1232(25)00192-4. [PMID: 40107353 DOI: 10.1016/j.jare.2025.03.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 02/19/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025] Open
Abstract
INTRODUCTION Minichromosome maintenance 4 (MCM4), a constituent of the MCM family, playing a pivotal role in DNA replication. Although MCM4 expression has been widely linked to various malignant tumors, its role in kidney diseases is not well-studied. This study primarily investigates the role and underlying mechanism of MCM4 in acute kidney injury (AKI). OBJECTIVES Characterizing a novel target of MCM4 in patients with AKI. METHODS We used CRISPR/Cas9 gene editing to delete MCM4 gene in tubular cells from C57BL/6J mice. Adeno-associated virus 9 harboring MCM4 was administered via intraparenchymal injection into the kidney to enhance MCM4 expression in vivo. These mice were used to established cisplatin- and ischemic reperfusion injury (IRI)-induced AKI mouse models, for detecting the functional role of MCM4 in the pathological process of AKI. RESULTS MCM4 level was increased in the tubules of cisplatin- and IRI-induced AKI mouse models. Compare to wide-type mice, MCM4 knockout mice demonstrated greater degree of histological damage and a higher ratio of apoptotic tubular cells, as well as kidney dysfunction upon cisplatin- and IRI-induced AKI models. Conversely, MCM4 overexpression ameliorated the severity of kidney injury and promoted regenerative capacity of tubular cells during AKI development. Mechanically, loss of MCM4 induced the expression of p53-binding protein 1, activating the p53/p21 pathway and exacerbating AKI progression. Additional, MAD2B, as an upstream molecule of MCM4, regulates the transcription level of MCM4 by affecting the level of E2F1. CONCLUSIONS These findings demonstrate that MCM4 upregulation during AKI development is an adaptive response that preserves tubular cell regenerative capacity and limits the severity of renal injury, thus highlighting the potential value of MCM4 as a biomarker or therapeutic target in patients with AKI.
Collapse
Affiliation(s)
- Jing Huang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Feng Liu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhi-Feng Xu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hui-Ling Xiang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qian Yuan
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
4
|
Srivastav AK, Jaiswal J, Rajput PK, Yadav UCS, Singh R, Kumar U. In silico and in vitro profiling of sphingomyelin loaded zein nanoconstructs with optimized aromatic interactions and pharmacokinetics for colon cancer therapeutics. Int J Biol Macromol 2025; 307:141845. [PMID: 40057096 DOI: 10.1016/j.ijbiomac.2025.141845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/25/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
To enhance therapeutic efficacy and achieve controlled drug release for colon cancer therapeutics, this study focuses on developing a hydrocolloid-based, pH-responsive biocompatible nanocarrier. We synthesized sphingomyelin (SM)-loaded zein nanoparticles (SM-LNPs) stabilized using 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC), leveraging the aromatic interactions of zein to optimize drug encapsulation and release kinetics. In silico molecular docking and molecular dynamics (MD) simulations demonstrated strong intermolecular interactions between zein, SM, and EDC, supporting the formulation's rational design. Comprehensive physicochemical characterization confirmed the stability and structural integrity of SM-LNPs. Dynamic light scattering (DLS) and field emission scanning electron microscopy (FE-SEM) analyses revealed a monodispersed, spherical morphology with an optimized particle size suitable for oral drug delivery. In vitro drug release studies indicated a sustained and pH-responsive release profile, with 90.60 % of SM released at pH 6.8, mimicking intestinal conditions. Cell viability assays using HCT 116 colon cancer cells showed that SM-LNPs exhibited significantly enhanced cytotoxicity (41.44 % reduction in cell viability) compared to free SM, confirming their potential for cancer therapeutics. Additionally, pharmacokinetic simulations validated the controlled release and systemic retention of SM, supporting its suitability for oral administration. Our findings demonstrate that SM-LNPs offer a promising drug delivery platform with enhanced stability, pH response release, and improved bioactivity, paving the way for their potential application in colon cancer treatment. This research provides critical insights into nanocarrier design, drug delivery kinetics, and biomaterial-based therapeutic strategies for precision oncology.
Collapse
Affiliation(s)
- Amit Kumar Srivastav
- School of Nano Sciences, Central University of Gujarat, Gandhinagar, India; Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Jyoti Jaiswal
- School of Nano Sciences, Central University of Gujarat, Gandhinagar, India; Department of Microbiology & Biotechnology, Gujarat University, Ahmedabad, Gujarat, India
| | - Pradeep Kumar Rajput
- Special Center for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Umesh C S Yadav
- Special Center for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Rajesh Singh
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, USA; Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Umesh Kumar
- School of Nano Sciences, Central University of Gujarat, Gandhinagar, India; Nutrition Biology Department, School of Interdisciplinary and Applied Sciences, Central University of Haryana, Mahendergarh, Haryana, India.
| |
Collapse
|
5
|
Lazovic A, Markovic BS, Corovic I, Markovic T, Andjelkovic M, Stojanovic B, Jovanovic I, Mitrovic M. Unlocking the molecular mechanisms of anticancer and immunomodulatory potentials of cariprazine in triple negative breast cancer. Biomed Pharmacother 2025; 184:117931. [PMID: 39978031 DOI: 10.1016/j.biopha.2025.117931] [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: 11/04/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 02/22/2025] Open
Abstract
Triple-negative breast cancer (TNBC), a highly invasive type of cancer, is difficult to treat due to insufficient specific targets and low survival rates. Current therapy often encounters drug resistance or relapse; thus, repurposing existing drugs could revolutionize cancer treatment. This study examined the anticancer effects of the antipsychotics Cariprazine (CAR), Olanzapine (OLZ), and Clozapine (CLZ), and the immunomodulatory potential of CAR, in vitro and in vivo in TNBC models. In vitro, CAR, OLZ, and CLZ significantly inhibited the proliferation of TNBC cells. This inhibition occurred via the induction of mitochondrial apoptosis, G0/G1 cell cycle arrest, and the suppression of autophagy, as evidenced by the down-regulation of Bcl-2, p62, and pAKT; the upregulation of Bax and active caspase 3; the decrease of ΔΨM; and the promotion of cytochrome c release. In addition, CAR inhibited MDA-MB-231 cells migration. In vivo, CAR inhibited tumor growth in the 4T1 xenograft model without causing adverse effects and resulted in the mRNA upregulation caspase 9, p53, p21, and Beclin-1. In addition, CAR influenced the immune response by promoting the production of proinflammatory cytokines TNF-α, IFN-γ, IL-17, and IL-1β and increasing the percentage of TNF-α+, IL-17+, IL-1β+, and IFN-γ+ CD3+ splenocytes. In conclusion, compared with other antipsychotics, 5-FU, and cisplatin, CAR exerted the most potent anticancer activity in TNBC in vitro and in vivo. This efficacy may be attributed to its ability to regulate apoptosis and autophagy, promote G0/G1 cell cycle arrest, and modulate antitumor immune response, suggesting its therapeutic potential in breast cancer.
Collapse
Affiliation(s)
- Aleksandar Lazovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, Svetozar Markovic 69, Kragujevac 34000, Serbia.
| | - Bojana Simovic Markovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozar Markovic 69, Kragujevac 34000, Serbia.
| | - Irfan Corovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozar Markovic 69, Kragujevac 34000, Serbia; General Hospital of Novi Pazar, Department of Internal Medicine, General Živković 1, Novi Pazar 36300, Serbia.
| | - Tijana Markovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Svetozar Markovic 69, Kragujevac 34000, Serbia.
| | - Marija Andjelkovic
- Department of Medical Biochemistry, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića, 69, Kragujevac 34000, Serbia.
| | - Bojan Stojanovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, Svetozar Markovic 69, Kragujevac 34000, Serbia.
| | - Ivan Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozar Markovic 69, Kragujevac 34000, Serbia.
| | - Marina Mitrovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozar Markovic 69, Kragujevac 34000, Serbia; Department of Medical Biochemistry, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića, 69, Kragujevac 34000, Serbia.
| |
Collapse
|
6
|
Koothradan S, Nayeem S, Elyas KK. PEGylated iron oxide-gold core-shell nanoparticles for tumor-targeted delivery of Rapamycin. 3 Biotech 2025; 15:23. [PMID: 39735611 PMCID: PMC11669639 DOI: 10.1007/s13205-024-04189-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/07/2024] [Indexed: 12/31/2024] Open
Abstract
Rapamycin analogs are approved by the FDA for breast and renal cancer treatment. Hence, the possibility of nanoparticle-mediated delivery of Rapamycin could be examined. In the present study, PEGylated Gold-core shell iron oxide nanoparticles were used for the targeted delivery of Rapamycin, and R-Au-IONPs were formulated. SEM, XRD, and FTIR determined the smooth spherical morphology, and compositional structure, and confirmed the conjugation of Rapamycin onto the NPs. The in vitro drug release study showed a controlled release of the drug over time. R-Au-IONPs showed significant cytotoxicity in MCF 7 cells. Anti-proliferative assays such as trypan blue dye exclusion assay, microscopy, Fluorescent staining, and clonogenic assays were performed. NH staining, Rhodamine 123 staining, PS externalization, and the cleavage of PARP protein by western immunoblot assays confirmed the induction of apoptosis. The mechanism of R-Au-IONP-induced cell death was analyzed by flow cytometry. Our in-vitro study, on the impact of R-Au-IONPs on cell viability in the human breast adenocarcinoma cell line (MCF-7), confirms the efficacy of drug delivery using the nanoparticle system. Further results implied the induction of apoptosis. This drug delivery system using Rapamycin could be a potential candidate in the treatment of breast cancer.
Collapse
Affiliation(s)
- Suhana Koothradan
- Department of Biotechnology, University of Calicut, Kerala Malappuram, 673635 India
| | - Safia Nayeem
- Department of Biotechnology, University of Calicut, Kerala Malappuram, 673635 India
| | - K. K. Elyas
- Department of Biotechnology, University of Calicut, Kerala Malappuram, 673635 India
| |
Collapse
|
7
|
Nadeem I, Han Z, Xiaoliang H, Adzraku SY, Kambey PA, Kanwore K, Peipei M, Adekunle AO, Adu-Amankwaah J, Ayanlaja AA, Zheng Y, Dianshuai G, Liu X, Song Y. Doublecortin regulates the mitochondrial-dependent apoptosis in glioma via Rho-A/Net-1/p38-MAPK signaling. Mol Med 2024; 30:272. [PMID: 39719558 DOI: 10.1186/s10020-024-01021-4] [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: 09/12/2024] [Accepted: 11/29/2024] [Indexed: 12/26/2024] Open
Abstract
Doublecortin (DCX) is a microtubule-associated protein known to be a key regulator of neuronal migration and differentiation during brain development. However, the role of DCX, particularly in regulating the survival and growth of glioma cells, remains unclear. In this study, we utilized CRISPR/Cas9 technology to knock down DCX in the human glioma cell line (U251). DCX depletion suppressed cell proliferation and enhanced the pro-apoptotic effects of temozolomide (TMZ) and γ-radiation treatment. DCX knockdown led to the translocation of Bax to the mitochondria and mitochondria dysfunction. Furthermore, DCX deficiency-induced apoptosis took place along with the generation of reactive oxygen species (ROS), which is crucial in triggering mitochondrial membrane depolarization, the release of cytochrome c (Cyt-c), and caspase activation. Importantly, the transcriptional inhibition of DCX downregulated Rho-A, Net-1, and activated p38-MAPK cue, critical for cell survival and proliferation. Subsequent treatment with TMZ and γ-radiation further increased p38-MAPK activity through the decreased expression of Rho-A/Net-1, resulting in a significant reduction in glioma cell migration and invasion. Additionally, intracranial xenograft tumors of DCX-modified U251 cells in nude mice demonstrated inhibited tumor growth. Tumor sections treated with TMZ and γ-radiation exhibited a higher number of TUNEL-positive cells compared to the control group, indicating increased apoptosis. Our finding suggests that DCX depletion reduces glioma cell proliferation and promotes mitochondria-dependent apoptosis by enhancing the chemo and radiotherapy response. Targeting DCX represents a potential therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Iqra Nadeem
- Department of Neurobiology and Anatomy, Key Laboratory of Neurobiology, Xuzhou Medical University, 209, Tongshan Road, Xuzhou, 221004, China
| | - Zhou Han
- Department of Neurobiology and Anatomy, Key Laboratory of Neurobiology, Xuzhou Medical University, 209, Tongshan Road, Xuzhou, 221004, China
| | - Hong Xiaoliang
- Department of Neurobiology and Anatomy, Key Laboratory of Neurobiology, Xuzhou Medical University, 209, Tongshan Road, Xuzhou, 221004, China
| | - Seyram Yao Adzraku
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002, China
| | - Piniel Alphayo Kambey
- Department of Neurobiology and Anatomy, Key Laboratory of Neurobiology, Xuzhou Medical University, 209, Tongshan Road, Xuzhou, 221004, China
| | - Kouminin Kanwore
- Department of Neurobiology and Anatomy, Key Laboratory of Neurobiology, Xuzhou Medical University, 209, Tongshan Road, Xuzhou, 221004, China
| | - Mu Peipei
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, China
| | | | - Joseph Adu-Amankwaah
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Abiola Abdulrahman Ayanlaja
- Department of Neurology, Johns Hopkins University School of Medicine, 201 N Broadway, Baltimore, MD, 21287, USA
| | - Yi Zheng
- Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Gao Dianshuai
- Department of Neurobiology and Anatomy, Key Laboratory of Neurobiology, Xuzhou Medical University, 209, Tongshan Road, Xuzhou, 221004, China.
| | - Xiaomei Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Yuanjian Song
- Department of Neurobiology and Anatomy, Key Laboratory of Neurobiology, Xuzhou Medical University, 209, Tongshan Road, Xuzhou, 221004, China.
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
8
|
Khiabani NA, Doustvandi MA, Story D, Nobari SA, Hajizadeh M, Petersen R, Dunbar G, Rossignol J. Glioblastoma therapy: State of the field and future prospects. Life Sci 2024; 359:123227. [PMID: 39537100 DOI: 10.1016/j.lfs.2024.123227] [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: 06/25/2024] [Revised: 09/03/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma (GB) is a cancerous brain tumor that originates from glial cells and leads to thousands of deaths each year and a five-year survival of only 6.8 %. Treatments for GB include surgery, chemotherapy, radiation, and immunotherapy. GB is an incurable fatal disease, necessitating the development of innovative strategies to find a developing effective therapy. Genetic therapies may be crucial in treating GB by identifying the mutations and amplifications of multiple genes, which drive its proliferation and spread. Use of small interfering RNAs (siRNAs) provides a novel technology used to suppress the genes associated with disease, which forms a basis for targeted therapy in GB and its stem cell population, which are recognized for their ability to develop resistance to chemotherapy and tumorigenic capabilities. This review examines the use of siRNAs in GB, emphasizing their effectiveness in suppressing key oncogenes and signaling pathways associated with tumor development, invasion, stemness, and resistance to standard treatments. siRNA-based gene silencing is a promising approach for developing targeted therapeutics against GB and associated stem cell populations, potentially enhancing patient outcomes and survival rates in this devastating disease.
Collapse
Affiliation(s)
- Nadia Allahyarzadeh Khiabani
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA; Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA; College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | | | - Darren Story
- Department of Psychology, Saginaw Valley State University, University Center, MI 48710, USA
| | | | | | - Robert Petersen
- College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | - Gary Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA; Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA; Department of Psychology, Central Michigan University, Mount Pleasant, MI, USA
| | - Julien Rossignol
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA; Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA; College of Medicine, Central Michigan University, Mount Pleasant, MI, USA.
| |
Collapse
|
9
|
Xiao QH, Xiang H, Tian YN, Huang JL, Li MQ, Wang PQ, Lian K, Yu PX, Xu MY, Zhang RN, Zhang Y, Huang J, Zhang WC, Duan P. Polystyrene microplastics alleviate the developmental toxicity of silver nanoparticles in embryo-larval zebrafish (Danio rerio) at the transcriptomic level. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176485. [PMID: 39341243 DOI: 10.1016/j.scitotenv.2024.176485] [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: 07/06/2024] [Revised: 09/12/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024]
Abstract
Since silver nanoparticles (AgNPs) and polystyrene microplastics (PS-MP) share common environmental niches, their interactions can modulate their hazard impacts. Herein, we assessed the developmental toxicity of 1 mg/L PS-MP, 0.5 mg/L AgNPs and the mixtures of AgNPs and PS-MP on embryo-larval zebrafish. We found that AgNPs co-exposure with PS-MP remarkably decreased mortality rates, malformation rates, heart rates and yolk sac area, while it increased hatching rates and eye size compared to the AgNPs group. These phenomena revealed that the cell cycle, oxidative stress, apoptosis, lipid metabolism, ferroptosis and p53 signalling pathway were obviously affected by single AgNPs exposure at 96 hpf (hours post fertilization). Interestingly, all these effects were effectively ameliorated by co-exposure with PS-MP. The combination of transcriptomic and metabolomic analyses showed that the imbalance of DEGs (differentially expressed genes) and DEMs (differentially expressed metabolites) (PI, phosphatidylinositol and TAG-FA, triacylglycerol-fatty acid) disturbed both the cell cycle and lipid metabolism following single AgNPs exposure and co-exposure with PS-MP. These findings suggest that PS-MP attenuates the developmental toxicity of AgNPs on embryo-larval zebrafish. Overall, this study provides important insight into understanding the transcriptional responses and mechanisms of AgNPs alone or in combination with PS-MPs on embryo-larval zebrafish, providing a reference for ecological risk assessment of combined exposure to PS-MP and metal nanoparticles.
Collapse
Affiliation(s)
- Qiao-Hong Xiao
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China; Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Hao Xiang
- Department of Nuclear Medicine, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Ya-Nan Tian
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China; Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Jiao-Long Huang
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China; Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Ming-Qun Li
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China; Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Pu-Qing Wang
- Hubei Provincial Clinical Research Center for Parkinson's Disease, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Kai Lian
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Peng-Xia Yu
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China; Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Meng-Yao Xu
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Ruo-Nan Zhang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Yan Zhang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Jie Huang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Wei-Cheng Zhang
- Center for Environment and Health in Water Source Area of South-to-North Water Diversion, School of Public Health, Hubei University of Medicine, Shiyan 442000, China.
| | - Peng Duan
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China; Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynaecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China.
| |
Collapse
|
10
|
Zarska M, Novak O, Jakubcova T, Novotny F, Urbancokova A, Havel F, Novak J, Raabova H, Musilek K, Filimonenko V, Bartek J, Proska J, Hodny Z. Photothermal induction of pyroptosis in malignant glioma spheroids using (16-mercaptohexadecyl)trimethylammonium bromide-modified cationic gold nanorods. Colloids Surf B Biointerfaces 2024; 243:114128. [PMID: 39094210 DOI: 10.1016/j.colsurfb.2024.114128] [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: 06/18/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
Plasmonic photothermal therapy (PPTT) employing plasmonic gold nanorods (GNRs) presents a potent strategy for eradication of tumors including aggressive brain gliomas. Despite its promise, there is a pressing need for a more comprehensive evaluation of PPTT using sophisticated in vitro models that closely resemble tumor tissues, thereby facilitating the elucidation of therapeutic mechanisms. In this study, we exposed 3D glioma spheroids (tumoroids) to (16-mercaptohexadecyl)trimethylammonium bromide-functionalized gold nanorods (MTAB-GNRs) and a near-infrared (NIR) laser. We demonstrate that the photothermal effect can be fine-tuned by adjusting the nanoparticle concentration and laser power. Depending on the selected parameters, the laser can trigger either regulated or non-regulated cell death (necrosis) in both mouse GL261 and human U-87 MG glioma cell lines, accompanied by translocation of phosphatidylserine in the membrane. Our investigation into the mechanism of regulated cell death induced by PPTT revealed an absence of markers associated with classical apoptosis pathways, such as cleaved caspase 3. Instead, we observed the presence of cleaved caspase 1, gasdermin D, and elevated levels of NLRP3 in NIR-irradiated tumoroids, indicating the activation of pyroptosis. This finding correlates with previous observations of lysosomal accumulation of MTAB-GNRs and the known lysosomal pathway of pyroptosis activation. We further confirmed the absence of toxic breakdown products of GNRs using electron microscopy, which showed no melting or fragmentation of gold nanoparticles under the conditions causing regulated cell death. In conclusion, PPTT using coated gold nanorods offers significant potential for glioma cell elimination occurring through the activation of pyroptosis rather than classical apoptosis pathways.
Collapse
Affiliation(s)
- Monika Zarska
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Ondrej Novak
- Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tereza Jakubcova
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Filip Novotny
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Alexandra Urbancokova
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Filip Havel
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Department of Laser Physics and Photonics, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Josef Novak
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Helena Raabova
- Electron Microscopy Core Facility, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Kamil Musilek
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic; Biomedical Research Center, University Hospital, Hradec Kralove, Czech Republic
| | - Vlada Filimonenko
- Electron Microscopy Core Facility, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Laboratory of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiri Bartek
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Genome Integrity Group, Danish Cancer Institute, Danish Cancer Society, Copenhagen, Denmark; Department of Medical Biochemistry and Biophysics, Science For Life Laboratory, Division of Genome Biology, Karolinska Institute, Stockholm, Sweden
| | - Jan Proska
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Department of Laser Physics and Photonics, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Zdenek Hodny
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
11
|
Molavand M, Ebrahimnezhade N, Kiani A, Yousefi B, Nazari A, Majidinia M. Regulation of autophagy by non-coding RNAs in human glioblastoma. Med Oncol 2024; 41:260. [PMID: 39375229 DOI: 10.1007/s12032-024-02513-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/18/2024] [Indexed: 10/09/2024]
Abstract
Glioblastoma, a lethal form of brain cancer, poses substantial challenges in treatment due to its aggressive nature and resistance to standard therapies like radiation and chemotherapy. Autophagy has a crucial role in glioblastoma progression by supporting cellular homeostasis and promoting survival under stressful conditions. Non-coding RNAs (ncRNAs) play diverse biological roles including, gene regulation, chromatin remodeling, and the maintenance of cellular homeostasis. Emerging evidence reveals the intricate regulatory mechanisms of autophagy orchestrated by non-coding RNAs (ncRNAs) in glioblastoma. The diverse roles of these ncRNAs in regulating crucial autophagy-related pathways, including AMPK/mTOR signaling, the PI3K/AKT pathway, Beclin1, and other autophagy-triggering system regulation, sheds light on ncRNAs biological mechanisms in the proliferation, invasion, and therapy response of glioblastoma cells. Furthermore, the clinical implications of targeting ncRNA-regulated autophagy as a promising therapeutic strategy for glioblastoma treatment are in the spotlight of ongoing studies. In this review, we delve into our current understanding of how ncRNAs regulate autophagy in glioblastoma, with a specific focus on microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), and their intricate interplay with therapy response.
Collapse
Affiliation(s)
- Mehran Molavand
- Student Research Commitee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Niloufar Ebrahimnezhade
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Arash Kiani
- Student Research Commite, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Bahman Yousefi
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran.
- Molecular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ahmad Nazari
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran.
- Tehran University of Medical Sciences, Tehran, Iran.
| | - Maryam Majidinia
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
12
|
Kaynar A, Kim W, Ceyhan AB, Zhang C, Uhlén M, Turkez H, Shoaie S, Mardinoglu A. Unveiling the Molecular Mechanisms of Glioblastoma through an Integrated Network-Based Approach. Biomedicines 2024; 12:2237. [PMID: 39457550 PMCID: PMC11504402 DOI: 10.3390/biomedicines12102237] [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: 09/06/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Despite current treatments extending the lifespan of Glioblastoma (GBM) patients, the average survival time is around 15-18 months, underscoring the fatality of GBM. This study aims to investigate the impact of sample heterogeneity on gene expression in GBM, identify key metabolic pathways and gene modules, and explore potential therapeutic targets. Methods: In this study, we analysed GBM transcriptome data derived from The Cancer Genome Atlas (TCGA) using genome-scale metabolic models (GEMs) and co-expression networks. We examine transcriptome data incorporating tumour purity scores (TPSs), allowing us to assess the impact of sample heterogeneity on gene expression profiles. We analysed the metabolic profile of GBM by generating condition-specific GEMs based on the TPS group. Results: Our findings revealed that over 90% of genes showing brain and glioma specificity in RNA expression demonstrate a high positive correlation, underscoring their expression is dominated by glioma cells. Conversely, negatively correlated genes are strongly associated with immune responses, indicating a complex interaction between glioma and immune pathways and non-tumorigenic cell dominance on gene expression. TPS-based metabolic profile analysis was supported by reporter metabolite analysis, highlighting several metabolic pathways, including arachidonic acid, kynurenine and NAD pathway. Through co-expression network analysis, we identified modules that significantly overlap with TPS-correlated genes. Notably, SOX11 and GSX1 are upregulated in High TPS, show a high correlation with TPS, and emerged as promising therapeutic targets. Additionally, NCAM1 exhibits a high centrality score within the co-expression module, which shows a positive correlation with TPS. Moreover, LILRB4, an immune-related gene expressed in the brain, showed a negative correlation and upregulated in Low TPS, highlighting the importance of modulating immune responses in the GBM mechanism. Conclusions: Our study uncovers sample heterogeneity's impact on gene expression and the molecular mechanisms driving GBM, and it identifies potential therapeutic targets for developing effective treatments for GBM patients.
Collapse
Affiliation(s)
- Ali Kaynar
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (A.K.); (A.B.C.); (S.S.)
| | - Woonghee Kim
- Science for Life Laboratory, KTH-Royal Institute of Technology, 171211 Stockholm, Sweden; (W.K.); (C.Z.); (M.U.)
| | - Atakan Burak Ceyhan
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (A.K.); (A.B.C.); (S.S.)
| | - Cheng Zhang
- Science for Life Laboratory, KTH-Royal Institute of Technology, 171211 Stockholm, Sweden; (W.K.); (C.Z.); (M.U.)
| | - Mathias Uhlén
- Science for Life Laboratory, KTH-Royal Institute of Technology, 171211 Stockholm, Sweden; (W.K.); (C.Z.); (M.U.)
| | - Hasan Turkez
- Medical Biology Department, Faculty of Medicine, Atatürk University, Erzurum 25240, Türkiye;
| | - Saeed Shoaie
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (A.K.); (A.B.C.); (S.S.)
- Science for Life Laboratory, KTH-Royal Institute of Technology, 171211 Stockholm, Sweden; (W.K.); (C.Z.); (M.U.)
| | - Adil Mardinoglu
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (A.K.); (A.B.C.); (S.S.)
- Science for Life Laboratory, KTH-Royal Institute of Technology, 171211 Stockholm, Sweden; (W.K.); (C.Z.); (M.U.)
| |
Collapse
|
13
|
Dong R, Meng X, Chang H, Lei Y, Hu Y, Yan Y, Song G. Titanium Dioxide Nanoparticles Induce Cell Cycle Arrest and Apoptosis through Inhibiting PI3K/AKT/mTOR Pathway in Spermatogonia. Biol Trace Elem Res 2024; 202:4065-4077. [PMID: 38079059 DOI: 10.1007/s12011-023-03984-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 11/27/2023] [Indexed: 07/18/2024]
Abstract
Titanium dioxide nanoparticles (TiO2 NPs) can result in the reduction of sperm numbers, but the mechanisms have not been well elucidated. The purpose of this study was to investigate the effects of TiO2 NPs on cell cycle and apoptosis in spermatogonia and to explore the role of PI3K/AKT/mTOR signaling pathway in this process. The mouse spermatogonia cell line (GC-1) was treated with TiO2 NPs at different concentrations (0, 25, 50, 75 and 100 μg/mL) for 24 h to detect cell viability, cell cycle, apoptosis, and key proteins related to cell cycle and PI3K/AKT/mTOR signaling pathway. The agonist (IGF-1) and inhibitor (LY294002) of PI3K were used to verify the role of PI3K/AKT/mTOR signaling pathway in cell cycle and apoptosis. TiO2 NPs significantly inhibited cell proliferation, induced cell cycle arrest at G0/G1 phase and resulted in apoptosis. TiO2 NPs downregulated the levels of cyclin-dependent kinases (CDKs) and cyclins, including CDK4, CDK2, Cyclin D1 and Cyclin E1, while upregulated the levels of p21 and p53 proteins. Furthermore, TiO2 NPs inhibited the PI3K/AKT/mTOR signaling pathway by decreasing the levels of p-PI3K, p-AKT and p-mTOR. IGF-1 reversed the G0/G1 phase arrest and apoptosis caused by TiO2 NPs. However, LY294002 aggravated the G0/G1 phase arrest and apoptosis resulting from TiO2 NPs. Collectively, TiO2 NPs induced cell cycle arrest at G0/G1 phase and apoptosis through inhibiting the activation of PI3K/AKT/mTOR pathway, which could be the main reason for the reduction in sperm numbers caused by TiO2 NPs.
Collapse
Affiliation(s)
- Ruoyun Dong
- Department of Preventive Medicine / the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Xiaojia Meng
- The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongmei Chang
- Department of Preventive Medicine / the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Yuzhu Lei
- Department of Preventive Medicine / the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Yunhua Hu
- Department of Preventive Medicine / the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Yizhong Yan
- Department of Preventive Medicine / the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Guanling Song
- Department of Preventive Medicine / the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China.
| |
Collapse
|
14
|
Madani F, Morovvati H, Webster TJ, Najaf Asaadi S, Rezayat SM, Hadjighassem M, Khosravani M, Adabi M. Combination chemotherapy via poloxamer 188 surface-modified PLGA nanoparticles that traverse the blood-brain-barrier in a glioblastoma model. Sci Rep 2024; 14:19516. [PMID: 39174603 PMCID: PMC11341868 DOI: 10.1038/s41598-024-69888-1] [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: 03/08/2024] [Accepted: 08/09/2024] [Indexed: 08/24/2024] Open
Abstract
The effect of chemotherapy for anti-glioblastoma is limited due to insufficient drug delivery across the blood-brain-barrier. Poloxamer 188-coated nanoparticles can enhance the delivery of nanoparticles across the blood-brain-barrier. This study presents the design, preparation, and evaluation of a combination of PLGA nanoparticles (PLGA NPs) loaded with methotrexate (P-MTX NPs) and PLGA nanoparticles loaded with paclitaxel (P-PTX NPs), both of which were surface-modified with poloxamer188. Cranial tumors were induced by implanting C6 cells in a rat model and MRI demonstrated that the tumors were indistinguishable in the two rats with P-MTX NPs + P-PTX NPs treated groups. Brain PET scans exhibited a decreased brain-to-background ratio which could be attributed to the diminished metabolic tumor volume. The expression of Ki-67 as a poor prognosis factor, was significantly lower in P-MTX NPs + P-PTX NPs compared to the control. Furthermore, the biodistribution of PLGA NPs was determined by carbon quantum dots loaded into PLGA NPs (P-CQD NPs), and quantitative analysis of ex-vivo imaging of the dissected organs demonstrated that 17.2 ± 0.6% of the NPs were concentrated in the brain after 48 h. The findings highlight the efficacy of combination nanochemotherapy in glioblastoma treatment, indicating the need for further preclinical studies.
Collapse
Affiliation(s)
- Fatemeh Madani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Morovvati
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Thomas J Webster
- School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, China
- Program in Materials Science, UFPI, Teresina, Brazil
| | - Sareh Najaf Asaadi
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoudreza Hadjighassem
- Brain and Spinal Cord Injury Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Masood Khosravani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Abd El-Lateef HM, Gaafar A, Alqahtani AS, Al-Mutairi AA, Alshaya DS, Elsaid FG, Fayad E, Farouk NA. Design, synthesis, and antiproliferative screening of new quinoline derivatives bearing a cis-vinyl triamide motif as apoptosis activators and EGFR-TK inhibitors. RSC Adv 2024; 14:24781-24790. [PMID: 39114435 PMCID: PMC11305403 DOI: 10.1039/d4ra04915b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
In this work, a congeneric set of quinoline-tethered cis-vinyl triamide hybrids was prepared and evaluated as EGFR tyrosine kinase inhibitors for the management of breast cancer. All of the prepared hybrids were evaluated for their antiproliferative effect against the breast MCF-7 cell line. Among the tested hybrids, compound 6f displayed the most potent antiproliferative activity with an IC50 value of 1.87 μM compared to STU (IC50 = 13.71 μM) as the standard reference. The most promising hybrid, 6f, was found to induce cellular cycle arrest at the G1 phase. Furthermore, the molecular mechanism of this hybrid revealed its ability to induce cellular apoptosis via the mitochondrial-dependent apoptotic pathway. Compound 6f decreased MCF-7 cells' MMP compared to the controls (percentage change value of 57.93%). Further investigation of the selective compound 6f showed that it can inhibit EGFR tyrosine kinase.
Collapse
Affiliation(s)
- Hany M Abd El-Lateef
- Department of Chemistry, College of Science, King Faisal University Al-Ahsa 31982 Saudi Arabia
- Department of Chemistry, Faculty of Science, Sohag University Sohag 82524 Egypt
| | - Ahmed Gaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Port Said University Port Said Egypt
| | - Arwa Sultan Alqahtani
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University(IMSIU) P.O. Box 90950 Riyadh 11623 Saudi Arabia
| | - Aamal A Al-Mutairi
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University(IMSIU) P.O. Box 90950 Riyadh 11623 Saudi Arabia
| | - Dalal Sulaiman Alshaya
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University P.O. Box 84428 Riyadh 11671 Saudi Arabia
| | - Fahmy Gad Elsaid
- Department of Biology, College of Science, King Khalid University PO Box 960 Abha 61421 Asir Saudi Arabia
| | - Eman Fayad
- Department of Biotechnology, College of Sciences, Taif University P.O. Box 11099 Taif 21944 Saudi Arabia
| | - N A Farouk
- Department of Chemistry, Faculty of Science, Port Said University Port Said Egypt
| |
Collapse
|
16
|
Liu J, Chang YT, Kou YY, Zhang PP, Dong QL, Guo RY, Liu LY, Lin HW, Yang F. Marine sponge-derived alkaloid inhibits the PI3K/AKT/mTOR signaling pathway against diffuse large B-cell lymphoma. Med Oncol 2024; 41:212. [PMID: 39073639 DOI: 10.1007/s12032-024-02448-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 07/10/2024] [Indexed: 07/30/2024]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a genetically heterogeneous non-Hodgkin lymphoma that is extremely aggressive and has an intermediate to high malignancy. Some patients still experience treatment failure, relapse, or resistance to rituximab, cyclophosphamide, adriamycin, vincristine, and prednisone (R-CHOP) therapy. Therefore, there is an urgent need for further research on new agents for the treatment of DLBCL. AP-48 is an aaptamine alkaloid analog with potent anti-tumor effects that originates from marine natural products. In this study, we found that AP-48 exhibits dose-dependent cytotoxicity in DLBCL cell lines. Flow cytometry showed that AP-48 induced cell cycle arrest in the G0/G1 phase in SU-DHL-4 and Farage cells and in the S phase in WSU-DLCL-2 cells. AP-48 also accelerated apoptosis via the caspase-3-mediated intrinsic apoptotic pathway. Further experiments demonstrated that AP-48 exerted its anti-DLBCL effects through the PI3K/AKT/mTOR pathway, and that the PI3K agonist YS49 partially alleviated the inhibition of cell proliferation and apoptosis induced by AP-48. Finally, in a tumor xenograft model, AP-48 inhibited tumor growth and promoted apoptosis in tumor tissues, indicating its therapeutic potential in DLBCL.
Collapse
Affiliation(s)
- Jie Liu
- Department of Pharmacy, Research Center for Marine Drugs, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yung-Ting Chang
- Department of Pharmacy, Research Center for Marine Drugs, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yan-Yu Kou
- Department of Pharmacy, Research Center for Marine Drugs, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Pei-Pei Zhang
- Department of Marine Biological Injury and Dermatology, Naval Special Medical Center, Naval Medical University, Shanghai, 200052, China.
| | - Qing-Li Dong
- School of Health Science and Engineering, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai, 200093, China
| | - Ruo-Yu Guo
- Key Laboratory of Marine Ecosystem Dynamics, Second Institute of Oceanography, Ministry of Natural Resources, 36 Baochubei Road, Hangzhou, 310012, China
| | - Li-Yun Liu
- Department of Pharmacy, Research Center for Marine Drugs, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Hou-Wen Lin
- Department of Pharmacy, Research Center for Marine Drugs, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Fan Yang
- Department of Pharmacy, Research Center for Marine Drugs, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
17
|
da Silva NM, Lopes ICS, Galué-Parra AJ, Ferreira IM, de Sena CBC, da Silva EO, Macchi BDM, de Oliveira FR, do Nascimento JLM. Fatty Acid Amides Suppress Proliferation via Cannabinoid Receptors and Promote the Apoptosis of C6 Glioma Cells in Association with Akt Signaling Pathway Inhibition. Pharmaceuticals (Basel) 2024; 17:873. [PMID: 39065724 PMCID: PMC11280372 DOI: 10.3390/ph17070873] [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: 04/15/2024] [Revised: 06/11/2024] [Accepted: 06/16/2024] [Indexed: 07/28/2024] Open
Abstract
A glioma is a type of tumor that acts on the Central Nervous System (CNS) in a highly aggressive manner. Gliomas can occasionally be inaccurately diagnosed and treatments have low efficacy, meaning that patients exhibit a survival of less than one year after diagnosis. Due to factors such as intratumoral cell variability, inefficient chemotherapy drugs, adaptive resistance development to drugs and tumor recurrence after resection, the search continues for new drugs that can inhibit glioma cell growth. As such, analogues of endocannabinoids, such as fatty acid amides (FAAs), represent interesting alternatives for inhibiting tumor growth, since FAAs can modulate several metabolic pathways linked to cancer and, thus, may hold potential for managing glioblastoma. The aim of this study was to investigate the in vitro effects of two fatty ethanolamides (FAA1 and FAA2), synthetized via direct amidation from andiroba oil (Carapa guianensis Aublet), on C6 glioma cells. FAA1 and FAA2 reduced C6 cell viability, proliferation and migratory potential in a dose-dependent manner and were not toxic to normal retina glial cells. Both FAAs caused apoptotic cell death through the loss of mitochondrial integrity (ΔΨm), probably by activating cannabinoid receptors, and inhibiting the PI3K/Akt pathway. In conclusion, FAAs derived from natural products may have the potential to treat glioma-type brain cancer.
Collapse
Affiliation(s)
- Nágila Monteiro da Silva
- Programa de Pós-Graduação em Neurociências e Biologia Celular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil; (N.M.d.S.); (I.C.S.L.); (E.O.d.S.)
- Laboratorio de Neuroquímica Molecular e Celular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil;
| | - Izabella Carla Silva Lopes
- Programa de Pós-Graduação em Neurociências e Biologia Celular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil; (N.M.d.S.); (I.C.S.L.); (E.O.d.S.)
- Laboratorio de Neuroquímica Molecular e Celular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil;
| | - Adan Jesus Galué-Parra
- Laboratório de Biologia Estrutural, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-750, Brazil; (A.J.G.-P.); (C.B.C.d.S.)
| | - Irlon Maciel Ferreira
- Laboratório de Biocatálise e Síntese Orgânica Aplicada, Departamento de Ciências Exatas e Tecnológicas, Universidade Federal do Amapá, Macapá 68902-280, Brazil;
- Programa de Pós-Graduação em Ciências Farmacêuticas, Departamento de Ciências Biológicas e da Saúde, Universidade Federal do Amapá, Macapá 68902-280, Brazil
| | - Chubert Bernardo Castro de Sena
- Laboratório de Biologia Estrutural, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-750, Brazil; (A.J.G.-P.); (C.B.C.d.S.)
- Instituto Nacional de Ciência e Tecnologia em Neuroimunomodulação (INCT-NIM), Rio de Janeiro 21040-900, Brazil
| | - Edilene Oliveira da Silva
- Programa de Pós-Graduação em Neurociências e Biologia Celular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil; (N.M.d.S.); (I.C.S.L.); (E.O.d.S.)
- Laboratório de Biologia Estrutural, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-750, Brazil; (A.J.G.-P.); (C.B.C.d.S.)
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem (INCT-INBEB), Rio de Janeiro 21941-902, Brazil
| | - Barbarella de Matos Macchi
- Laboratorio de Neuroquímica Molecular e Celular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Neuroimunomodulação (INCT-NIM), Rio de Janeiro 21040-900, Brazil
- Programa de Pós-Graduação em Farmacologia e Bioquímica, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil
| | - Fábio Rodrigues de Oliveira
- Laboratório de Controle de Qualidade e Bromatologia, Curso de Farmácia, Departamento de Ciências Biológicas e da Saúde, Universidade Federal do Amapá, Macapá 68902-280, Brazil;
| | - José Luiz Martins do Nascimento
- Programa de Pós-Graduação em Neurociências e Biologia Celular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil; (N.M.d.S.); (I.C.S.L.); (E.O.d.S.)
- Laboratorio de Neuroquímica Molecular e Celular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil;
- Programa de Pós-Graduação em Ciências Farmacêuticas, Departamento de Ciências Biológicas e da Saúde, Universidade Federal do Amapá, Macapá 68902-280, Brazil
- Instituto Nacional de Ciência e Tecnologia em Neuroimunomodulação (INCT-NIM), Rio de Janeiro 21040-900, Brazil
- Programa de Pós-Graduação em Farmacologia e Bioquímica, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil
| |
Collapse
|
18
|
Dixon S, O'connor AT, Brooks-Noreiga C, Clark MA, Levy A, Castejon AM. Role of renin angiotensin system inhibitors and metformin in Glioblastoma Therapy: a review. Cancer Chemother Pharmacol 2024; 94:1-23. [PMID: 38914751 DOI: 10.1007/s00280-024-04686-0] [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] [Accepted: 06/13/2024] [Indexed: 06/26/2024]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive and incurable disease accounting for about 10,000 deaths in the USA each year. Despite the current treatment approach which includes surgery with chemotherapy and radiation therapy, there remains a high prevalence of recurrence. Notable improvements have been observed in persons receiving concurrent antihypertensive drugs such as renin angiotensin inhibitors (RAS) or the antidiabetic drug metformin with standard therapy. Anti-tumoral effects of RAS inhibitors and metformin have been observed in in vitro and in vivo studies. Although clinical trials have shown mixed results, the potential for the use of RAS inhibitors and metformin as adjuvant GBM therapy remains promising. Nevertheless, evidence suggest that these drugs exert multimodal antitumor actions; by particularly targeting several cancer hallmarks. In this review, we highlight the results of clinical studies using multidrug cocktails containing RAS inhibitors and or metformin added to standard therapy for GBM. In addition, we highlight the possible molecular mechanisms by which these repurposed drugs with an excellent safety profile might elicit their anti-tumoral effects. RAS inhibition elicits anti-inflammatory, anti-angiogenic, and immune sensitivity effects in GBM. However, metformin promotes anti-migratory, anti-proliferative and pro-apoptotic effects mainly through the activation of AMP-activated protein kinase. Also, we discussed metformin's potential in targeting both GBM cells as well as GBM associated-stem cells. Finally, we summarize a few drug interactions that may cause an additive or antagonistic effect that may lead to adverse effects and influence treatment outcome.
Collapse
Affiliation(s)
- Sashana Dixon
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Ft. Lauderdale, FL, USA.
| | - Ann Tenneil O'connor
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Chloe Brooks-Noreiga
- Halmos College of Arts and Sciences, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Michelle A Clark
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Arkene Levy
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Ana M Castejon
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Ft. Lauderdale, FL, USA
| |
Collapse
|
19
|
Nguyen TTT, Greene LA, Mnatsakanyan H, Badr CE. Revolutionizing Brain Tumor Care: Emerging Technologies and Strategies. Biomedicines 2024; 12:1376. [PMID: 38927583 PMCID: PMC11202201 DOI: 10.3390/biomedicines12061376] [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: 05/09/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive forms of brain tumor, characterized by a daunting prognosis with a life expectancy hovering around 12-16 months. Despite a century of relentless research, only a select few drugs have received approval for brain tumor treatment, largely due to the formidable barrier posed by the blood-brain barrier. The current standard of care involves a multifaceted approach combining surgery, irradiation, and chemotherapy. However, recurrence often occurs within months despite these interventions. The formidable challenges of drug delivery to the brain and overcoming therapeutic resistance have become focal points in the treatment of brain tumors and are deemed essential to overcoming tumor recurrence. In recent years, a promising wave of advanced treatments has emerged, offering a glimpse of hope to overcome the limitations of existing therapies. This review aims to highlight cutting-edge technologies in the current and ongoing stages of development, providing patients with valuable insights to guide their choices in brain tumor treatment.
Collapse
Affiliation(s)
- Trang T. T. Nguyen
- Ronald O. Perelman Department of Dermatology, Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, NY 10016, USA
| | - Lloyd A. Greene
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA;
| | - Hayk Mnatsakanyan
- Department of Neurology, Massachusetts General Hospital, Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA; (H.M.); (C.E.B.)
| | - Christian E. Badr
- Department of Neurology, Massachusetts General Hospital, Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA; (H.M.); (C.E.B.)
| |
Collapse
|
20
|
Abdelrahman Z, Abdelatty A, Luo J, McKnight AJ, Wang X. Stratification of glioma based on stemness scores in bulk and single-cell transcriptomes. Comput Biol Med 2024; 175:108304. [PMID: 38663352 DOI: 10.1016/j.compbiomed.2024.108304] [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: 10/28/2023] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 05/15/2024]
Abstract
BACKGROUND Brain tumours are known to have a high mortality and morbidity rate due to their localised and frequent invasive growth. The concept that glioma resistance could originate from the dissimilarity in the vulnerability of clonogenic glial stem cells to chemotherapeutic drugs and radiation has driven the scientific community to reexamine the comprehension of glioma growth and strategies that target these cells or modify their stemness. METHODS Based on the enrichment scores of 12 stemness signatures, we identified glioma subtypes in both tumour bulks and single cells by clustering analysis. Furthermore, we comprehensively compared molecular and clinical features among the glioma subtypes. RESULTS Consistently, in seven different datasets, hierarchical clustering uncovered three subtypes of glioma, termed Stem-H, Stem-M, and Stem-L, with high, medium, and low stemness signatures, respectively. Stem-H and Stem-L exhibited the most unfavorable and favourable overall and disease-free survival, respectively. Stem-H showed the highest enrichment scores of the EMT, invasion, proliferation, differentiation, and metastasis processes signatures, while Stem-L displayed the lowest. Stem-H harboured a greater proportion of late-stage tumours compared to Stem-L. Moreover, Stem-H manifested higher tumour mutation burden, DNA damage repair and cell cycle activity, intratumour heterogeneity, and a more frequent incidence of TP53 and EGFR mutations than Stem-L. In contrast, Stem-L had higher O6-Methylguanine-DNA Methyltransferase (MGMT) methylation levels. CONCLUSION The classification of glioma based on stemness may offer new insights into the biology of the tumour, as well as more accurate clinical management of the disease.
Collapse
Affiliation(s)
- Zeinab Abdelrahman
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, BT12 6BA, UK.
| | - Alaa Abdelatty
- Department of Pathology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Jiangti Luo
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Amy Jayne McKnight
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, BT12 6BA, UK
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
21
|
Emami Meybodi SM, Moradi Moraddahande F, Dehghani Firoozabadi A. Immunogenic cell death mediated TLR3/4-activated MSCs in U87 GBM cell line. Heliyon 2024; 10:e29858. [PMID: 38698968 PMCID: PMC11064142 DOI: 10.1016/j.heliyon.2024.e29858] [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: 02/17/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/05/2024] Open
Abstract
Background and aims Glioblastoma (GBM) is an aggressive primary brain cancer with no promising curative therapies. It has been indicated that MSCs can interact with the tumour microenvironment (TME) through the secretion of soluble mediators regulating intercellular signalling within the TME. TLRs are a multigene family of pattern recognition receptors with evolutionarily conserved regions and are widely expressed in immune and other body cells. MSCs by TLRs can recognize conserved molecular components (DAPMPs and PAPMPs) and activate signalling pathways, which regulate immune and inflammatory responses. MSCs may exert immunomodulatory functions through interaction with their expressed toll-like receptors (TLRs) and exert a protective effect against tumour antigens. As an emerging approach, we aimed to monitor the U87 cell line growth, migration and death markers following specific TLR3/4-primed-MSCs-CMs treatment. Methods and results We investigated the phenotypic and functional outcomes of primed-CMs and glioma cell line co-culture following short-term, low-dose TLR3/4 priming. The gene expression profile of target genes, including apoptotic markers and related genes, was analyzed by qRT-PCR. MicroRNA-Seq examined the miRNA expression patterns, and flow cytometry evaluated the cell viability and cycle stages. The results showed significant changes in apoptosis and likely necroptosis-related markers following TLR3/4-primed-MSCs-CMs exposure in the glioma cell line. Notably, we observed a considerable induction of selective pro-apoptotic markers and both the early and late stages of apoptosis in treated U87 cell lines. Additionally, the migration rate of glioma cells significantly decreased following MSCs-CM treatment. Conclusion Our findings confirmed that the exposure of TLR3/4-activated-MSCs-CMs with glioma tumour cells possibly changes the immunogenicity of the tumour microenvironment and induces immunogenic programmed cell death. Our results can support the idea that TLR3/4-primed-MSCs can lead to innate immune-mediated cell death and modify tumour cell biology in invasive and metastatic cancers.
Collapse
Affiliation(s)
- Seyed Mahdi Emami Meybodi
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Moradi Moraddahande
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Dehghani Firoozabadi
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
22
|
Valerius AR, Webb LM, Sener U. Novel Clinical Trials and Approaches in the Management of Glioblastoma. Curr Oncol Rep 2024; 26:439-465. [PMID: 38546941 DOI: 10.1007/s11912-024-01519-4] [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] [Accepted: 03/14/2024] [Indexed: 05/02/2024]
Abstract
PURPOSE OF REVIEW The purpose of this review is to discuss a wide variety of novel therapies recently studied or actively undergoing study in patients with glioblastoma. This review also discusses current and future strategies for improving clinical trial design in patients with glioblastoma to maximize efficacy in discovering effective treatments. RECENT FINDINGS Over the years, there has been significant expansion in therapy modalities studied in patients with glioblastoma. These therapies include, but are not limited to, targeted molecular therapies, DNA repair pathway targeted therapies, immunotherapies, vaccine therapies, and surgically targeted radiotherapies. Glioblastoma is the most common malignant primary brain tumor in adults and unfortunately remains with poor overall survival following the current standard of care. Given the dismal prognosis, significant clinical and research efforts are ongoing with the goal of improving patient outcomes and enhancing quality and quantity of life utilizing a wide variety of novel therapies.
Collapse
Affiliation(s)
| | - Lauren M Webb
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Ugur Sener
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
23
|
Chamani R, Saberi O, Fathinejad F. An arresten-derived anti-angiogenic peptide triggers apoptotic cell death in endothelial cells. Mol Biol Rep 2024; 51:513. [PMID: 38622345 DOI: 10.1007/s11033-024-09448-y] [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: 11/01/2023] [Accepted: 03/13/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND In recent years, anti-angiogenic peptides have received considerable attention as candidates for cancer treatment. Arresten is an angiogenesis inhibitor that cleaves from the α1 chain of type IV collagen and stimulates apoptosis in endothelial cells. We have recently indicated that a peptide corresponding to the amino acid 78 to 86 of arresten, so-called Ars, prevented the migration and tube formation of HUVECs and the colon carcinoma growth in mice significantly. The current study aimed to determine whether induction of apoptotic cell death in endothelial cells is one of the biochemical mechanisms of this anti-angiogenic peptide. METHODS AND RESULTS This hypothesis was assessed using the MTT assay, cell cycle analysis, Annexin V-FITC/PI staining, BCL2, CASP8, CASP9, p53, and CDKN2A gene expression studies as well as evaluating apoptosis in tumor tissues by TUNEL assay. Results demonstrated that 40 µM of Ars significantly stimulated 46.2% of early and late apoptosis in HUVECs compared to 13.6% in the untreated cells and did not significantly alter the cell cycle distribution. Moreover, BCL2 and CASP8 were down-regulated, while CASP9 and p53 were up-regulated in endothelial cells. CDKN2A gene expression, the regulator of G1 cell cycle arrest, was not significantly altered. CONCLUSIONS It might be suggested that Ars induced apoptosis in endothelial cells through the mitochondrial pathway and had no effect on the cell cycle. Besides, Ars induced apoptosis significantly in vivo. However, further studies are required to confirm the detailed molecular mechanism of Ars, this peptide has the potential to be optimized for clinical translations.
Collapse
Affiliation(s)
| | - Omid Saberi
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Fatemeh Fathinejad
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| |
Collapse
|
24
|
Jacobs J, Iranpour R, Behrooz AB, da Silva Rosa SC, Ghavami S. The role of BCL2L13 in glioblastoma: turning a need into a target. Biochem Cell Biol 2024; 102:127-134. [PMID: 37988705 DOI: 10.1139/bcb-2023-0221] [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] [Indexed: 11/23/2023] Open
Abstract
Glioblastoma (GBM) is the most common aggressive central nervous system cancer. GBM has a high mortality rate, with a median survival time of 12-15 months after diagnosis. A poor prognosis and a shorter life expectancy may result from resistance to standard treatments such as radiation and chemotherapy. Temozolomide has been the mainstay treatment for GBM, but unfortunately, there are high rates of resistance with GBM bypassing apoptosis. A proposed mechanism for bypassing apoptosis is decreased ceramide levels, and previous research has shown that within GBM cells, B cell lymphoma 2-like 13 (BCL2L13) can inhibit ceramide synthase. This review aims to discuss the causes of resistance in GBM cells, followed by a brief description of BCL2L13 and an explanation of its mechanism of action. Further, lipids, specifically ceramide, will be discussed concerning cancer and GBM cells, focusing on ceramide synthase and its role in developing GBM. By gathering all current information on BCL2L13 and ceramide synthase, this review seeks to enable an understanding of these pieces of GBM in the hope of finding an effective treatment for this disease.
Collapse
Affiliation(s)
- Joadi Jacobs
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Rosa Iranpour
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Simone C da Silva Rosa
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Research Institute of Hematology and Oncology, Cancer Care Manitoba, Winnipeg, MB R3E 0V9, Canada
- Faculty of Medicine in Zabrze, University of Technology in Katowice, Academia of Silesia, 41-800 Zabrze, Poland
| |
Collapse
|
25
|
Zou J, Zhang H, Wu Z, Hu W, Zhang T, Xie H, Huang Y, Zhou H. TIGD1 Is an Independent Prognostic Factor that Promotes the Progression of Colon Cancer. Cancer Biother Radiopharm 2024; 39:223-235. [PMID: 36508261 DOI: 10.1089/cbr.2022.0052] [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: 12/14/2022] Open
Abstract
Background: Trigger transposable element-derived 1 (TIGD1) is a human-specific gene, but no studies have been conducted to determine its mechanism of action. Our aim is to ascertain the function and mode of action of TIGD1 in the development of colon cancer. Materials and Methods: The authors used bioinformatics to analyze the relationship between TIGD1 and the clinical characteristics of colon cancer, as well as its prognosis. A series of cell assays were conducted to assess the function of TIGD1 in the proliferation and migration of colon cancer, and flow cytometry was used to explore its effects on apoptosis and the cell cycle. Results: The authors discovered that the expression of TIGD1 was remarkably elevated in colon cancer. Clinical correlation analysis demonstrated that TIGD1 expression was elevated in the tissues of advanced-stage patients, and it was remarkably elevated in individuals with both lymph node and distant metastasis. Further, the authors found that individuals showing elevated TIGD1 expression levels had a shortened survival time. Univariate and multivariate Cox regression analyses revealed that TIGD1 was an independent prognostic factor. Overexpression of the TIGD1 gene remarkedly enhances the proliferation and metastasis of colon cancer cells and suppresses apoptosis. In addition, the overexpression of TIGD1 can enhance the transition of tumor cells from the G1 toward the S phase. Western blot results suggested that TIGD1 may promote the malignant activity of colon cancer cells via the Wnt/β-catenin signaling pathway, Bcl-2, N-cadherin, BAX, E-cadherin, CDK6, and CyclinD1. Conclusions: TIGD1 may be an independent prognostic factor in the advancement of colon cancer, and therefore function as a therapeutic target.
Collapse
Affiliation(s)
- Junwei Zou
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Hesong Zhang
- Department of Hepatobiliary Surgery, The Second People's Hospital of Wuhu, Wuhu, China
| | - Zhaoying Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Weichao Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Tingting Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Hao Xie
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Yong Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Hailang Zhou
- Department of Gastroenterology, Lianshui County People's Hospital, Huai'an, China
| |
Collapse
|
26
|
Wang Z, Lu X, Liu C, Huang F, Lu T, Chen Y, Liu L, Lu S. Discovery of FLT3-targeting PROTACs with potent antiproliferative activity against acute myeloid leukemia cells harboring FLT3 mutations. Eur J Med Chem 2024; 268:116237. [PMID: 38387337 DOI: 10.1016/j.ejmech.2024.116237] [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: 01/13/2024] [Revised: 02/02/2024] [Accepted: 02/12/2024] [Indexed: 02/24/2024]
Abstract
Acute myeloid leukemia (AML) patients harboring Fms-like tyrosine kinase 3 (FLT3) mutations often suffer from poor prognosis and relapse. Targeted protein degradation utilizing proteolysis targeting chimeras (PROTACs) is considered as a novel therapeutic strategy in drug discovery and may be a promising modality to target FLT3 mutations for the development of potent anti-AML drugs. Herein, a kind of FLT3-targeting PROTACs was rationally developed based on a FLT3 inhibitor previously reported by us. The representative compound 35 showed potent and selective antiproliferative activities against AML cells harboring FLT3 mutations. Western blot assay demonstrated that compound 35 effectively induced the degradation of FLT3-ITD and decreased the phosphorylation levels of FLT3-ITD, AKT, STAT5 and ERK in MV4-11 cells in a dose-dependent manner. Flow cytometry analysis illustrated that compound 35 strongly induced apoptosis and cell cycle arrest in MV4-11 cells in a dose-dependent manner. Moreover, compound 35 displayed favorable metabolic stability in in-vitro liver microsomes studies. Comparative molecular dynamic (MD) simulation studies further elucidated the underlying mechanism of compound 35 to stabilize the dynamic ensemble of the FLT3-compound 35-cereblon (CRBN) ternary complex. Taken together, compound 35 could serve as a lead molecule for developing FLT3 degraders against AML.
Collapse
Affiliation(s)
- Zhijie Wang
- ShenZhen Hospital, Southern Medical University, Shenzhen, 518000, PR China; School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Xun Lu
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Canlin Liu
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Fei Huang
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Tao Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Lifei Liu
- Department of Infectious Disease, Children's Hospital of Nanjing Medical University, Nanjing, 210008, PR China.
| | - Shuai Lu
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China.
| |
Collapse
|
27
|
Nkuimi OBK, Silihe KK, Tabi YO, Ngo Pambe JC, Njamen D, Zingue S. Duguetia confinis Engl. & Diels (Annonaceae) inhibitory and cytotoxic effects on breast adenocarcinoma growth both in vitro and in vivo. Heliyon 2024; 10:e24410. [PMID: 38317996 PMCID: PMC10839804 DOI: 10.1016/j.heliyon.2024.e24410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/30/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Background Breast cancer is ranked as the most common malignant tumor in women globally with ∼ 2.3 million new cases (11.7 %) diagnosed in 2020. The multiple drawbacks associated with treatments, prompt researchers and patients to search for alternative therapy. Plants continue to offer encouraging leads, in particular those of the Annonaceae family, to which belongs Duguetia confinis, used by Cameroonian traditional healers to fight cancers. This study was aimed at investigating the effect of Duguetia confinis against human breast cancer cells. This was carried out by investigating the cytotoxicity, underlying mechanism of action and chemopreventive potential of D. confinis on 7,12-dimethylbenz(a)anthracene (DMBA)-induced breast cancer. Methods To achieve this goal, the ethanolic extract of the bark of D. confinis was prepared and assayed for its ability to inhibit cell growth, cell proliferation and clone formation. Furthermore, cell death mechanisms, cell cycle progression and anti-metastatic potential were investigated. The in vivo study consisted in a once-off administration of 50 mg/kg BW DMBA (in olive oil, s.c) from the 10th day after pretreatment with D. confinis extract (50 and 100 mg/kg BW) or standards [tamoxifen (3.3 mg/kg) and letrozole (1 mg/kg)] or leaf extract of Annona muricata L. (200 mg/kg as pharmacological control). Normal and negative controls received vehicle (3 % ethanol). The treatment of animals was done for 20 weeks, followed by the assessment of the incidence, burden and volume of tumors, breast cancer biomarker (CA 15-3), antioxidant status, inflammatory status and histopathology profile. The LD50 of D. confinis extract was estimated according to OECD guideline 423. Results D. confinis displayed cytotoxicity at 80 μg/mL on all the tested breast cancer cell lines. It induced apoptosis and caused a blockade at G0/G1; S-phase of MDA-MB 231 cells, thus, suggesting anticancer potential. A significant concentration-dependent antimetastatic potential was observed with D. confinis extract at 50 (p < 0.05) and 100 (p < 0.01) μg/mL, evidenced by a reduction in cell migration, chemotaxis and increased adhesion to extracellular matrix. With respect to the chemopreventive study, D. confinis was able to prevent the onset of breast adenocarcinoma in Wistar rats by preventing the growth of tumor mass and volume, as well as the histopathological severity of the disease. This was achieved through the modulation of antioxidant parameters (SOD, CAT, MDA) and inflammatory parameters (IL-12, IL-6, INF- gamma, TNF). Also, the LD50 of D. confinis extract was greater than 2000 mg/kg, indicating low acute toxicity and thus, favorable for therapeutic use. Conclusion In summary, this study outlines for the first time the beneficial effect of D. confinis as a plant candidate in the fight against breast cancer just like other species of the Annonaceae family. However, further research studies are still warranted regarding its bioactive components, and in depth investigation of its anticancer mechanism of action are also needed.
Collapse
Affiliation(s)
- Ornella Bernie Kami Nkuimi
- Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| | - Kevine Kamga Silihe
- Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
- Department of Pharmacotoxicology and Pharmacokinetics, Faculty of Medicine and Biomedical Sciences, University of Yaounde 1, P.O. Box 1364, Yaoundé, Cameroon
| | - Yves Omgba Tabi
- Department of Pharmacotoxicology and Pharmacokinetics, Faculty of Medicine and Biomedical Sciences, University of Yaounde 1, P.O. Box 1364, Yaoundé, Cameroon
| | - Judith Christiane Ngo Pambe
- Department of Morphological Sciences and Pathological Anatomy, Faculty of Medicine and Biomedical Sciences, University of Garoua, P.O. Box 317, Garoua, Cameroon
| | - Dieudonné Njamen
- Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| | - Stéphane Zingue
- Department of Pharmacotoxicology and Pharmacokinetics, Faculty of Medicine and Biomedical Sciences, University of Yaounde 1, P.O. Box 1364, Yaoundé, Cameroon
| |
Collapse
|
28
|
Navone SE, Guarnaccia L, Rizzaro MD, Begani L, Barilla E, Alotta G, Garzia E, Caroli M, Ampollini A, Violetti A, Gervasi N, Campanella R, Riboni L, Locatelli M, Marfia G. Role of Luteolin as Potential New Therapeutic Option for Patients with Glioblastoma through Regulation of Sphingolipid Rheostat. Int J Mol Sci 2023; 25:130. [PMID: 38203299 PMCID: PMC10779390 DOI: 10.3390/ijms25010130] [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: 10/25/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor, still considered incurable. In this study, conducted on primary GBM stem cells (GSCs), specifically selected as the most therapy-resistant, we examined the efficacy of luteolin, a natural flavonoid, as an anti-tumoral compound. Luteolin is known to impact the sphingolipid rheostat, a pathway regulated by the proliferative sphingosine-1-phosphate (S1P) and the proapoptotic ceramide (Cer), and implicated in numerous oncopromoter biological processes. Here, we report that luteolin is able to inhibit the expression of SphK1/2, the two kinases implicated in S1P formation, and to increase the expression of both SGPL1, the lyase responsible for S1P degradation, and CERS1, the ceramide synthase 1, thus shifting the balance toward the production of ceramide. In addition, luteolin proved to decrease the expression of protumoral signaling as MAPK, RAS/MEK/ERK and PI3K/AKT/mTOR and cyclins involved in cell cycle progression. In parallel, luteolin succeeded in upregulation of proapoptotic mediators as caspases and Bcl-2 family and cell cycle controllers as p53 and p27. Furthermore, luteolin determined the shutdown of autophagy contributing to cell survival. Overall, our data support the use of luteolin as add-on therapy, having demonstrated a good ability in impairing GSC viability and survival and increasing cell sensitivity to TMZ.
Collapse
Affiliation(s)
- Stefania Elena Navone
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Massimiliano D. Rizzaro
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Laura Begani
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Emanuela Barilla
- Andremacon Biotech Srl, Viale Ortles, 22/4, 20141 Milan, Italy; (E.B.); (G.A.); (R.C.); (L.R.)
| | - Giovanni Alotta
- Andremacon Biotech Srl, Viale Ortles, 22/4, 20141 Milan, Italy; (E.B.); (G.A.); (R.C.); (L.R.)
| | - Emanuele Garzia
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 Milan, Italy;
- Aerospace Medicine Institute “A. Mosso”, Italian Air Force, 20138 Milan, Italy
| | - Manuela Caroli
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Antonella Ampollini
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Aniello Violetti
- Space Attache’, Embassy of Italy in Washington DC, Washington, DC 20008, USA
| | - Noreen Gervasi
- Alcamena Stem Cell Therapeutics, 1450 South Rolling Road, Suite 4.069, Halethorpe, MD 21227, USA
| | - Rolando Campanella
- Andremacon Biotech Srl, Viale Ortles, 22/4, 20141 Milan, Italy; (E.B.); (G.A.); (R.C.); (L.R.)
| | - Laura Riboni
- Andremacon Biotech Srl, Viale Ortles, 22/4, 20141 Milan, Italy; (E.B.); (G.A.); (R.C.); (L.R.)
| | - Marco Locatelli
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
- Department of Medical-Surgical Physiopathology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
- Aerospace Medicine Institute “A. Mosso”, Italian Air Force, 20138 Milan, Italy
| |
Collapse
|
29
|
Jabir MS, Al-Shammari AM, Ali ZO, Albukhaty S, Sulaiman GM, Jawad SF, Hamzah SS, Syed A, Elgorban AM, Eswaramoorthy R, Zaghloul NSS, Al-Dulimi AG, Najm MAA. Combined oncolytic virotherapy gold nanoparticles as synergistic immunotherapy agent in breast cancer control. Sci Rep 2023; 13:16843. [PMID: 37803068 PMCID: PMC10558528 DOI: 10.1038/s41598-023-42299-4] [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: 02/28/2023] [Accepted: 09/07/2023] [Indexed: 10/08/2023] Open
Abstract
Combining viruses and nanoparticles may be a way to successfully treat cancer and minimize adverse effects. The current work aimed to evaluate the efficacy of a specific combination of gold nanoparticles (GNPs) and Newcastle disease virus (NDV) to enhance the antitumor effect of breast cancer in both in vitro and in vivo models. Two human breast cancer cell lines (MCF-7 and AMJ-13) and a normal epithelial cell line (HBL-100) were used and treated with NDV and/or GNPs. The MTT assay was used to study the anticancer potentials of NDV and GNP. The colony formation assay and apoptosis markers were used to confirm the killing mechanisms of NDV and GNP against breast cancer cell lines. p53 and caspase-9 expression tested by the qRT-PCR technique. Our results showed that combination therapy had a significant killing effect against breast cancer cells. The findings demonstrated that NDV and GNPs induced apoptosis in cancer cells by activating caspase-9, the p53 protein, and other proteins related to apoptosis, which holds promise as a combination therapy for breast cancer.
Collapse
Affiliation(s)
- Majid S Jabir
- Division of Biotechnology, Department of Applied Sciences, University of Technology, Baghdad, 10066, Iraq.
| | - Ahmed M Al-Shammari
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetics Research, Mustansiriyah University, Baghdad, Iraq.
| | - Zainab O Ali
- Division of Biotechnology, Department of Applied Sciences, University of Technology, Baghdad, 10066, Iraq
| | - Salim Albukhaty
- Department of Chemistry, College of Science, University of Misan, Maysan, 62001, Iraq
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq
| | - Ghassan M Sulaiman
- Division of Biotechnology, Department of Applied Sciences, University of Technology, Baghdad, 10066, Iraq.
| | - Sabrean F Jawad
- Department of Pharmacy, Al-Mustaqbal University College, Babylon, Iraq
| | - Sawsan S Hamzah
- College of Dentistry, Department of Basic Sciences, Ibn Sina University of Medical and Pharmaceutical Sciences, Baghdad, Iraq
| | - Asad Syed
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. 2455, 11451, Riyadh, Saudi Arabia
| | - Abdallah M Elgorban
- Center of Excellence in Biotechnology Research, King Saud University, Riyadh, Saudi Arabia
| | - Rajalakshmanan Eswaramoorthy
- Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, 600 077, India
| | - Nouf S S Zaghloul
- Bristol Centre for Functional Nanomaterials, HH Wills Physics Laboratory, Tyndall Avenue, University of Bristol, Bristol, BS8 1FD, UK
| | - Ali G Al-Dulimi
- Department of Dentistry, Bilad Alrafidain University College, Diyala, 32001, Iraq
| | - Mazin A A Najm
- Pharmaceutical Chemistry Department, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| |
Collapse
|
30
|
Rastegar-Pouyani N, Montazeri V, Marandi N, Aliebrahimi S, Andalib M, Jafarzadeh E, Montazeri H, Ostad SN. The Impact of Cancer-Associated Fibroblasts on Drug Resistance, Stemness, and Epithelial-Mesenchymal Transition in Bladder Cancer: A Comparison between Recurrent and Non-Recurrent Patient-Derived CAFs. Cancer Invest 2023; 41:656-671. [PMID: 37462514 DOI: 10.1080/07357907.2023.2237576] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/14/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
This study comparatively evaluated the possible effects of recurrent and non-recurrent patient-derived Cancer-Associated Fibroblasts (CAFs-R and -NR) on the bladder cancer cell line, EJ138. Both groups of CAFs increased cisplatin resistance and altered cell cycle distribution alongside induced resistance to apoptosis. Later, the scratch assay confirmed the cell migration-inducing effects of CAFs on cells. Nonetheless, only CAFs-R managed to increase sphere-formation and clonogenic levels in EJ138 cells, which were later validated by upregulating pluripotency transcription factors. Besides, CAFs-R also affected the expression levels of some of the EMT markers. Our study suggests that CAFs-R had stronger pro-tumorigenic effects on EJ138 cells.
Collapse
Affiliation(s)
- Nima Rastegar-Pouyani
- Department of Pharmacology and Toxicology, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahideh Montazeri
- Department of Artificial Intelligence, Smart University of Medical Sciences, Tehran, Iran
| | - Nikoo Marandi
- School of Pharmacy, Islamic Azad University of Medical Sciences, Tehran Iran
| | - Shima Aliebrahimi
- Department of Artificial Intelligence, Smart University of Medical Sciences, Tehran, Iran
| | - Melika Andalib
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Emad Jafarzadeh
- Department of Pharmacology and Toxicology, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Montazeri
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Nasser Ostad
- Department of Pharmacology and Toxicology, Tehran University of Medical Sciences, Tehran, Iran
- Toxicology and Poisoning Research Centre, Department of Toxicology and Pharmacology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Tluli O, Al-Maadhadi M, Al-Khulaifi AA, Akomolafe AF, Al-Kuwari SY, Al-Khayarin R, Maccalli C, Pedersen S. Exploring the Role of microRNAs in Glioma Progression, Prognosis, and Therapeutic Strategies. Cancers (Basel) 2023; 15:4213. [PMID: 37686489 PMCID: PMC10486509 DOI: 10.3390/cancers15174213] [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: 06/26/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 09/10/2023] Open
Abstract
Gliomas, which arise from glial cells in the brain, remain a significant challenge due to their location and resistance to traditional treatments. Despite research efforts and advancements in healthcare, the incidence of gliomas has risen dramatically over the past two decades. The dysregulation of microRNAs (miRNAs) has prompted the creation of therapeutic agents that specially target them. However, it has been reported that they are involved in complex signaling pathways that contribute to the loss of expression of tumor suppressor genes and the upregulation of the expression of oncogenes. In addition, numerous miRNAs promote the development, progression, and recurrence of gliomas by targeting crucial proteins and enzymes involved in metabolic pathways such as glycolysis and oxidative phosphorylation. However, the complex interplay among these pathways along with other obstacles hinders the ability to apply miRNA targeting in clinical practice. This highlights the importance of identifying specific miRNAs to be targeted for therapy and having a complete understanding of the diverse pathways they are involved in. Therefore, the aim of this review is to provide an overview of the role of miRNAs in the progression and prognosis of gliomas, emphasizing the different pathways involved and identifying potential therapeutic targets.
Collapse
Affiliation(s)
- Omar Tluli
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (O.T.); (M.A.-M.); (A.A.A.-K.); (A.F.A.); (R.A.-K.)
| | - Mazyona Al-Maadhadi
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (O.T.); (M.A.-M.); (A.A.A.-K.); (A.F.A.); (R.A.-K.)
| | - Aisha Abdulla Al-Khulaifi
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (O.T.); (M.A.-M.); (A.A.A.-K.); (A.F.A.); (R.A.-K.)
| | - Aishat F. Akomolafe
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (O.T.); (M.A.-M.); (A.A.A.-K.); (A.F.A.); (R.A.-K.)
| | - Shaikha Y. Al-Kuwari
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (O.T.); (M.A.-M.); (A.A.A.-K.); (A.F.A.); (R.A.-K.)
| | - Roudha Al-Khayarin
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (O.T.); (M.A.-M.); (A.A.A.-K.); (A.F.A.); (R.A.-K.)
| | | | - Shona Pedersen
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (O.T.); (M.A.-M.); (A.A.A.-K.); (A.F.A.); (R.A.-K.)
| |
Collapse
|
32
|
Hellmold D, Kubelt C, Daunke T, Beckinger S, Janssen O, Hauck M, Schütt F, Adelung R, Lucius R, Haag J, Sebens S, Synowitz M, Held-Feindt J. Sequential Treatment with Temozolomide Plus Naturally Derived AT101 as an Alternative Therapeutic Strategy: Insights into Chemoresistance Mechanisms of Surviving Glioblastoma Cells. Int J Mol Sci 2023; 24:ijms24109075. [PMID: 37240419 DOI: 10.3390/ijms24109075] [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: 02/27/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Glioblastoma (GBM) is a poorly treatable disease due to the fast development of tumor recurrences and high resistance to chemo- and radiotherapy. To overcome the highly adaptive behavior of GBMs, especially multimodal therapeutic approaches also including natural adjuvants have been investigated. However, despite increased efficiency, some GBM cells are still able to survive these advanced treatment regimens. Given this, the present study evaluates representative chemoresistance mechanisms of surviving human GBM primary cells in a complex in vitro co-culture model upon sequential application of temozolomide (TMZ) combined with AT101, the R(-) enantiomer of the naturally occurring cottonseed-derived gossypol. Treatment with TMZ+AT101/AT101, although highly efficient, yielded a predominance of phosphatidylserine-positive GBM cells over time. Analysis of the intracellular effects revealed phosphorylation of AKT, mTOR, and GSK3ß, resulting in the induction of various pro-tumorigenic genes in surviving GBM cells. A Torin2-mediated mTOR inhibition combined with TMZ+AT101/AT101 partly counteracted the observed TMZ+AT101/AT101-associated effects. Interestingly, treatment with TMZ+AT101/AT101 concomitantly changed the amount and composition of extracellular vesicles released from surviving GBM cells. Taken together, our analyses revealed that even when chemotherapeutic agents with different effector mechanisms are combined, a variety of chemoresistance mechanisms of surviving GBM cells must be taken into account.
Collapse
Affiliation(s)
- Dana Hellmold
- Department of Neurosurgery, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Carolin Kubelt
- Department of Neurosurgery, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Tina Daunke
- Institute of Experimental Cancer Research, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Silje Beckinger
- Institute of Experimental Cancer Research, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Ottmar Janssen
- Institute for Immunology, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Margarethe Hauck
- Functional Nanomaterials, Department of Materials Science, Kiel University, 24143 Kiel, Germany
| | - Fabian Schütt
- Functional Nanomaterials, Department of Materials Science, Kiel University, 24143 Kiel, Germany
| | - Rainer Adelung
- Functional Nanomaterials, Department of Materials Science, Kiel University, 24143 Kiel, Germany
| | - Ralph Lucius
- Institute of Anatomy, Kiel University, 24098 Kiel, Germany
| | - Jochen Haag
- Department of Pathology, Kiel University, 24105 Kiel, Germany
| | - Susanne Sebens
- Institute of Experimental Cancer Research, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Michael Synowitz
- Department of Neurosurgery, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Janka Held-Feindt
- Department of Neurosurgery, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, 24105 Kiel, Germany
| |
Collapse
|
33
|
d-limonene-loaded liposomes target malignant glioma cells via the downregulation of angiogenic growth factors. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2023]
|
34
|
Exploring Core Genes by Comparative Transcriptomics Analysis for Early Diagnosis, Prognosis, and Therapies of Colorectal Cancer. Cancers (Basel) 2023; 15:cancers15051369. [PMID: 36900162 PMCID: PMC10000172 DOI: 10.3390/cancers15051369] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers with a high mortality rate. Early diagnosis and therapies for CRC may reduce the mortality rate. However, so far, no researchers have yet investigated core genes (CGs) rigorously for early diagnosis, prognosis, and therapies of CRC. Therefore, an attempt was made in this study to explore CRC-related CGs for early diagnosis, prognosis, and therapies. At first, we identified 252 common differentially expressed genes (cDEGs) between CRC and control samples based on three gene-expression datasets. Then, we identified ten cDEGs (AURKA, TOP2A, CDK1, PTTG1, CDKN3, CDC20, MAD2L1, CKS2, MELK, and TPX2) as the CGs, highlighting their mechanisms in CRC progression. The enrichment analysis of CGs with GO terms and KEGG pathways revealed some crucial biological processes, molecular functions, and signaling pathways that are associated with CRC progression. The survival probability curves and box-plot analyses with the expressions of CGs in different stages of CRC indicated their strong prognostic performance from the earlier stage of the disease. Then, we detected CGs-guided seven candidate drugs (Manzamine A, Cardidigin, Staurosporine, Sitosterol, Benzo[a]pyrene, Nocardiopsis sp., and Riccardin D) by molecular docking. Finally, the binding stability of four top-ranked complexes (TPX2 vs. Manzamine A, CDC20 vs. Cardidigin, MELK vs. Staurosporine, and CDK1 vs. Riccardin D) was investigated by using 100 ns molecular dynamics simulation studies, and their stable performance was observed. Therefore, the output of this study may play a vital role in developing a proper treatment plan at the earlier stages of CRC.
Collapse
|
35
|
Ortiz-Rivera J, Nuñez R, Kucheryavykh Y, Kucheryavykh L. The PYK2 inhibitor PF-562271 enhances the effect of temozolomide on tumor growth in a C57Bl/6-Gl261 mouse glioma model. J Neurooncol 2023; 161:593-604. [PMID: 36790653 PMCID: PMC9992029 DOI: 10.1007/s11060-023-04260-3] [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: 01/09/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND The development of resistance to temozolomide (TMZ), a standard chemotherapeutic, limits the effective treatment of glioblastoma (GBM). Focal adhesion kinase (FAK) and proline rich tyrosine kinase 2 (Pyk2) regulate proliferation and invasion of GBM cells. We found that TMZ activates FAK and Pyk2 signaling in GBM. We hypothesized that pharmacological inhibitors of Pyk2/FAK together with TMZ can enhance the inhibitory effect of TMZ on tumor growth and dispersal and improve the treatment outcome. METHODS Primary human GBM cell cultures and a C57Bl/6-GL261 mouse glioma implantation model were used. Pyk2 (Tyr579/580) and FAK (Tyr925) phosphorylation was analyzed by western blotting. Viability, cell cycle, migration, invasion and invadopodia formation were investigated in vitro. Animal survival, tumor size and invasion, TUNEL apoptotic cell death and the Ki67 proliferation index were evaluated in vivo upon treatment with TMZ (50 mg/kg, once/day, orally) and the Pyk2/FAK inhibitor PF-562271 (once/daily, 50 mg/kg, orally) vs. TMZ monotherapy. RESULTS In vitro studies revealed significantly reduced viability, cell cycle progression, invasion and invadopodia with TMZ (100 µM) + PF-562271 (16 nM) compared with TMZ alone. In vivo studies demonstrated that combinatorial treatment led to prominent reductions in tumor size and invasive margins, extensive signs of apoptosis and a reduced proliferation index, together with a 15% increase in the survival rate in animals, compared with TMZ monotherapy. CONCLUSION TMZ + PF-562271 eliminates TMZ-related Pyk2/FAK activation in GBM and improves the treatment efficacy.
Collapse
Affiliation(s)
- Jescelica Ortiz-Rivera
- Department of Biochemistry, School of Medicine, Universidad Central de Caribe, Bayamon, PR 00956 USA
| | - Rebeca Nuñez
- Department of Biochemistry, School of Medicine, Universidad Central de Caribe, Bayamon, PR 00956 USA
| | - Yuriy Kucheryavykh
- Department of Biochemistry, School of Medicine, Universidad Central de Caribe, Bayamon, PR 00956 USA
| | - Lilia Kucheryavykh
- Department of Biochemistry, School of Medicine, Universidad Central de Caribe, Bayamon, PR 00956 USA
| |
Collapse
|
36
|
Zhao MH, Liu W, Zhang X, Zhang Y, Luo B. Epstein-Barr virus miR-BART2-5p and miR-BART11-5p regulate cell proliferation, apoptosis, and migration by targeting RB and p21 in gastric carcinoma. J Med Virol 2023; 95:e28338. [PMID: 36418188 DOI: 10.1002/jmv.28338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/12/2022] [Accepted: 11/12/2022] [Indexed: 11/25/2022]
Abstract
Epstein-Barr virus (EBV) was the first tumor virus discovered in humans and can cause various types of tumors. Molecular classification suggests that EBV-associated gastric cancer (EBVaGC) is a unique subtype of gastric cancer.EBV was also the first virus found to encode its own microRNAs. However, the functions of many miRNAs remain unknown. This study investigated the roles and targets of miR-BART2-5p (BART2-5p) and miR-BART11-5p (BART11-5p) in EBVaGC. The expression of RB and p21 in EBVaGC and EBV negative GC (EBVnGC) cells was evaluated by western blotting. Expression of BART2-5p and BART11-5p in EBVaGC cells was evaluated by droplet digital PCR. The effects of BART2-5p or BART11-5p and their potential mechanisms were further investigated using cell counting kit-8, colony formation assay, flow cytometry analysis, and transwell assay. BART2-5p and BART11-5p were abundantly expressed and RB and p21 were downregulated in EBVaGC cells. BART2-5p regulates RB and p21 expression by directly targeting them. BART11-5p regulates RB expression by directly targeting RB. Both BART2-5p and BART11-5p promoted proliferation and migration of gastric cancer cells, while inhibiting apoptosis and promoting S-phase arrest of the cell cycle. Thus, BART2-5p and BART11-5p play important roles in promoting proliferation and migration, and inhibiting apoptosis in EBVaGC by targeting RB and p21, thus providing new potential therapeutic targets for EBVaGC.
Collapse
Affiliation(s)
- Meng-He Zhao
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wen Liu
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xing Zhang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yan Zhang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China.,Department of Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Bing Luo
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
37
|
Mowforth OD, Brannigan J, El Khoury M, Sarathi CIP, Bestwick H, Bhatti F, Mair R. Personalised therapeutic approaches to glioblastoma: A systematic review. Front Med (Lausanne) 2023; 10:1166104. [PMID: 37122327 PMCID: PMC10140534 DOI: 10.3389/fmed.2023.1166104] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Glioblastoma is the most common and malignant primary brain tumour with median survival of 14.6 months. Personalised medicine aims to improve survival by targeting individualised patient characteristics. However, a major limitation has been application of targeted therapies in a non-personalised manner without biomarker enrichment. This has risked therapies being discounted without fair and rigorous evaluation. The objective was therefore to synthesise the current evidence on survival efficacy of personalised therapies in glioblastoma. Methods Studies reporting a survival outcome in human adults with supratentorial glioblastoma were eligible. PRISMA guidelines were followed. MEDLINE, Embase, Scopus, Web of Science and the Cochrane Library were searched to 5th May 2022. Clinicaltrials.gov was searched to 25th May 2022. Reference lists were hand-searched. Duplicate title/abstract screening, data extraction and risk of bias assessments were conducted. A quantitative synthesis is presented. Results A total of 102 trials were included: 16 were randomised and 41 studied newly diagnosed patients. Of 5,527 included patients, 59.4% were male and mean age was 53.7 years. More than 20 types of personalised therapy were included: targeted molecular therapies were the most studied (33.3%, 34/102), followed by autologous dendritic cell vaccines (32.4%, 33/102) and autologous tumour vaccines (10.8%, 11/102). There was no consistent evidence for survival efficacy of any personalised therapy. Conclusion Personalised glioblastoma therapies remain of unproven survival benefit. Evidence is inconsistent with high risk of bias. Nonetheless, encouraging results in some trials provide reason for optimism. Future focus should address target-enriched trials, combination therapies, longitudinal biomarker monitoring and standardised reporting.
Collapse
Affiliation(s)
- Oliver D. Mowforth
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, England, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, England, United Kingdom
| | - Jamie Brannigan
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, England, United Kingdom
| | - Marc El Khoury
- School of Clinical Medicine, University of Cambridge, Cambridge, England, United Kingdom
| | | | - Harry Bestwick
- School of Clinical Medicine, University of Cambridge, Cambridge, England, United Kingdom
| | - Faheem Bhatti
- School of Clinical Medicine, University of Cambridge, Cambridge, England, United Kingdom
| | - Richard Mair
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, England, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, England, United Kingdom
- *Correspondence: Richard Mair,
| |
Collapse
|
38
|
Filippone A, Casili G, Scuderi SA, Mannino D, Lanza M, Campolo M, Paterniti I, Capra AP, Colarossi C, Bonasera A, Lombardo SP, Cuzzocrea S, Esposito E. Sodium Propionate Contributes to Tumor Cell Growth Inhibition through PPAR-γ Signaling. Cancers (Basel) 2022; 15:cancers15010217. [PMID: 36612214 PMCID: PMC9818202 DOI: 10.3390/cancers15010217] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/26/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
New therapeutic approaches are needed to improve the outcome of patients with glioblastoma (GBM). Propionate, a short-chain fatty acid (SCFA), has a potent antiproliferative effect on various tumor cell types. Peroxisome proliferator-activated receptor (PPAR) ligands possess anticancer properties. We aimed to investigate the PPAR-γ/SCFAs interaction in in vitro and in vivo models of GBM. The U87 cell line was used in the in vitro study and was treated with sodium propionate (SP). U87 cells were silenced by using PPAR-γ siRNA or Ctr siRNA. In the in vivo study, BALB/c nude mice were inoculated in the right flank with 3 × 106 U-87 cells. SP (doses of 30 and 100 mg/kg) and GW9662 (1 mg/kg) were administered. In vitro exposure of GBM to SP resulted in prominent apoptosis activation while the autophagy pathway was promoted by SP treatments by influencing autophagy-related proteins. Knockdown of PPAR-γ sensitized GBM cells and blocked the SP effect. In vivo, SP was able to decrease tumor growth and to resolve GBM tissue features. SP promoted apoptosis and autophagy pathways and tumor cell proliferation leading to cell cycle arrest through a PPAR-γ-dependent mechanism suggesting that the PPAR-γ/SCFAs axis could be targeted for the management of GBM.
Collapse
Affiliation(s)
- Alessia Filippone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D׳Alcontres, 31-98166 Messina, Italy
| | - Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D׳Alcontres, 31-98166 Messina, Italy
| | - Sarah Adriana Scuderi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D׳Alcontres, 31-98166 Messina, Italy
| | - Deborah Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D׳Alcontres, 31-98166 Messina, Italy
| | - Marika Lanza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D׳Alcontres, 31-98166 Messina, Italy
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D׳Alcontres, 31-98166 Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D׳Alcontres, 31-98166 Messina, Italy
| | - Anna Paola Capra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D׳Alcontres, 31-98166 Messina, Italy
| | - Cristina Colarossi
- Istituto Oncologico del Mediterraneo, Via Penninazzo, 7-95029 Catania, Italy
| | - Annalisa Bonasera
- Istituto Oncologico del Mediterraneo, Via Penninazzo, 7-95029 Catania, Italy
| | | | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D׳Alcontres, 31-98166 Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D׳Alcontres, 31-98166 Messina, Italy
- Correspondence: ; Tel.: +39-090-6765208
| |
Collapse
|
39
|
Sevastre AS, Manea EV, Popescu OS, Tache DE, Danoiu S, Sfredel V, Tataranu LG, Dricu A. Intracellular Pathways and Mechanisms of Colored Secondary Metabolites in Cancer Therapy. Int J Mol Sci 2022; 23:ijms23179943. [PMID: 36077338 PMCID: PMC9456420 DOI: 10.3390/ijms23179943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 12/03/2022] Open
Abstract
Despite the great advancements made in cancer treatment, there are still many unsatisfied aspects, such as the wide palette of side effects and the drug resistance. There is an obvious increasing scientific attention towards nature and what it can offer the human race. Natural products can be used to treat many diseases, of which some plant products are currently used to treat cancer. Plants produce secondary metabolites for their signaling mechanisms and natural defense. A variety of plant-derived products have shown promising anticancer properties in vitro and in vivo. Rather than recreating the natural production environment, ongoing studies are currently setting various strategies to significantly manipulate the quantity of anticancer molecules in plants. This review focuses on the recently studied secondary metabolite agents that have shown promising anticancer activity, outlining their potential mechanisms of action and pathways.
Collapse
Affiliation(s)
- Ani-Simona Sevastre
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 200349 Craiova, Romania
| | - Elena Victoria Manea
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 200349 Craiova, Romania
| | - Oana Stefana Popescu
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 200349 Craiova, Romania
| | - Daniela Elise Tache
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 200349 Craiova, Romania
| | - Suzana Danoiu
- Department of Pathophysiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 200349 Craiova, Romania
| | - Veronica Sfredel
- Department of Physiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 200349 Craiova, Romania
| | - Ligia Gabriela Tataranu
- Neurosurgical Department, Clinical Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
- Correspondence: ; Tel.: +40-21-334-30-25
| | - Anica Dricu
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 200349 Craiova, Romania
| |
Collapse
|
40
|
Lu J, Yu C, Bao Q, Zhang X, Wang J. Identification and analysis of necroptosis-associated signatures for prognostic and immune microenvironment evaluation in hepatocellular carcinoma. Front Immunol 2022; 13:973649. [PMID: 36081504 PMCID: PMC9445885 DOI: 10.3389/fimmu.2022.973649] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/05/2022] [Indexed: 12/18/2022] Open
Abstract
Background Hepatocellular carcinoma remains the third most common cause of cancer-related deaths worldwide. Although great achievements have been made in resection, chemical therapies and immunotherapies, the pathogenesis and mechanism of HCC initiation and progression still need further exploration. Necroptosis genes have been reported to play an important role in HCC malignant activities, thus it is of great importance to comprehensively explore necroptosis-associated genes in HCC. Methods We chose the LIHC cohort from the TCGA, ICGC and GEO databases for this study. ConsensusClusterPlus was adopted to identify the necroptosis genes-based clusters, and LASSO cox regression was applied to construct the prognostic model based on necroptosis signatures. The GSEA and CIBERSORT algorithms were applied to evaluate the immune cell infiltration level. QPCR was also applied in this study to evaluate the expression level of genes in HCC. Results We identified three clusters, C1, C2 and C3. Compared with C2 and C3, the C1 cluster had the shortest overall survival time and highest immune score. The C1 was samples were significantly enriched in cell cycle pathways, some tumor epithelial-mesenchymal transition related signaling pathways, among others. The DEGs between the 3 clusters showed that C1 was enriched in cell cycle, DNA replication, cellular senescence, and p53 signaling pathways. The LASSO cox regression identified KPNA2, SLC1A5 and RAMP3 as prognostic model hub genes. The high risk-score subgroup had an elevated expression level of immune checkpoint genes and a higher TIDE score, which suggested that the high risk-score subgroup had a lower efficiency of immunotherapies. We also validated that the necroptosis signatures-based risk-score model had powerful prognosis prediction ability. Conclusion Based on necroptosis-related genes, we classified patients into 3 clusters, among which C1 had significantly shorter overall survival times. The proposed necroptosis signatures-based prognosis prediction model provides a novel approach in HCC survival prediction and clinical evaluation.
Collapse
Affiliation(s)
- Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Juan Lu,
| | - Chengbo Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiongling Bao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoqian Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|