1
|
Cao S, Lun S, Duan L, Gao Z, Wang X, Li Y, Zhang Y. Harnessing Calmodulin-Related Genes to Build a Prognostic Model in Esophageal Squamous Cell Carcinoma for a Comprehensive Analysis of Single-Cell Immune Characteristics and Drug Efficacy. J Immunother 2025:00002371-990000000-00140. [PMID: 40375794 DOI: 10.1097/cji.0000000000000561] [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: 01/14/2025] [Accepted: 03/27/2025] [Indexed: 05/18/2025]
Abstract
SUMMARY Calmodulin (CALM) has a bearing on the prognosis of various cancers. However, the prognostic value of CALM in esophageal squamous cell carcinoma (ESCC) remains unelucidated. Differentially expressed genes (DEGs) were screened between normal and tumor groups of TCGA-ESCC sets. The intersection of DEGs with calmodulin-related genes (CRGs) yielded differentially expressed CRGs (DE-CRGs). A prognostic model was established using LASSO Cox regression analysis and multivariate Cox regression analysis. qPCR validated the expression of prognostic feature genes. Analysis of gene expression patterns of different cellular clusters was based on single-cell sequencing data. Lastly, GSEA enrichment, immune infiltration, mutational profiling, drug sensitivity, and molecular docking as well as cellular thermal shift assay (CETSA) were conducted for ESCC patients. A prognosis model with excellent predictive capability was created based on 4 feature genes (ATP2B3, CALB1, KCNQ1, and MYO1G). The qPCR results demonstrated that ATP2B3 and KCNQ1 were significantly downregulated in human ESCC cells, whereas CALB1 and MYO1G were upregulated (P<0.05). Single-cell analysis uncovered that MYO1G and KCNQ1 were mainly expressed in different cell clusters. Furthermore, this risk model was strongly associated with functional pathway enrichment, immune cell infiltration, and somatic mutations. We also identified AZD-8055 may be potential therapy for ESCC patients. The CETSA experiment demonstrated the existence of a favorable binding thermal stability between AZD-8055 and MYO1G. This research may identify potential biomarkers for predicting the prognosis of ESCC patients.
Collapse
Affiliation(s)
- Shasha Cao
- Henan Medical Key Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, China
| | | | | | | | | | | | | |
Collapse
|
2
|
Kulkarni GC, Saha R, Peters CJ. Ion channel expression and function in glioblastoma multiforme (GBM): pathophysiological mechanisms and therapeutic potential. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119982. [PMID: 40328081 DOI: 10.1016/j.bbamcr.2025.119982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/29/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025]
Abstract
Glioblastoma Multiforme (GBM) is a highly malignant and diffusely invasive WHO Grade IV brain tumor arising from glial and neural stem cells. GBM is characterized by rapid proliferation and migration, aggressive invasion of local brain parenchyma, a hypoxic microenvironment, resistance to apoptosis and high vascular remodeling and angiogenesis. These hallmarks contribute to a near universal tumor recurrence after treatment or resection and poor patient prognosis. Ion channels, a superfamily of proteins responsible for permitting ion flux across otherwise impermeant membranes, show extensive remodeling in GBM with aberrant function mechanistically linked to manipulation of each of these hallmarks. In this review, we will discuss the known links between ion channel expression and activity and cellular processes that are enhanced or perturbed during GBM formation or progression. We will also discuss the extent to which basic or translational findings on ion channels in GBM samples or cell lines have shown preclinical promise towards the development of improved therapeutics against GBMs.
Collapse
Affiliation(s)
- Gauri C Kulkarni
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, USA
| | - Rayna Saha
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, USA
| | - Christian J Peters
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
3
|
De Silva MI, Gan HK, Bardy C. Repurposing trifluoperazine for glioblastoma treatment. Trends Pharmacol Sci 2025; 46:392-406. [PMID: 40300936 DOI: 10.1016/j.tips.2025.03.005] [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: 02/10/2025] [Revised: 03/16/2025] [Accepted: 03/16/2025] [Indexed: 05/01/2025]
Abstract
Glioblastoma (GBM) remains a therapeutic challenge due to its heterogeneity and plasticity, which drive treatment resistance, especially when compounded by interactions with the brain microenvironment. Recent preclinical evidence indicates that trifluoperazine (TFP) inhibits treatment-induced malignant reprogramming of tumour cells, potentially helping to reduce tumour plasticity. TFP targets calmodulin, dopamine receptors, and stress-responsive proteins (nuclear protein 1, NUPR1). Through these mechanisms, TFP has been shown to reduce tumour growth, sensitise tumours to chemoradiotherapy, and prolong survival in xenograft animal models. The clinical safety profile of TFP is well known from its use as an antipsychotic, and recent preclinical evidence further indicates that TFP has low toxicity to healthy neurons and glia despite transient functional effects on dopamine receptors. This Opinion explores TFP mechanisms of action and clinical activity to assess its suitability as a repurposed therapeutic option for GBM.
Collapse
Affiliation(s)
- Manam Inushi De Silva
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; South Australian Health and Medical Research Institute (SAHMRI), Laboratory for Human Neurophysiology and Genetics, Adelaide, SA, Australia
| | - Hui K Gan
- Cancer Therapies and Biology Group, Centre of Research Excellence in Brain Tumours, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, VIC, Australia; La Trobe University School of Cancer Medicine, and Department of Medicine, University of Melbourne, Heidelberg, Melbourne, VIC, Australia
| | - Cedric Bardy
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; South Australian Health and Medical Research Institute (SAHMRI), Laboratory for Human Neurophysiology and Genetics, Adelaide, SA, Australia.
| |
Collapse
|
4
|
Anwer MS, Abdel-Rasol MA, El-Sayed WM. Emerging therapeutic strategies in glioblastsoma: drug repurposing, mechanisms of resistance, precision medicine, and technological innovations. Clin Exp Med 2025; 25:117. [PMID: 40223032 PMCID: PMC11994545 DOI: 10.1007/s10238-025-01631-0] [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: 02/16/2025] [Accepted: 03/11/2025] [Indexed: 04/15/2025]
Abstract
Glioblastoma (GBM) is an aggressive Grade IV brain tumor with a poor prognosis. It results from genetic mutations, epigenetic changes, and factors within the tumor microenvironment (TME). Traditional treatments like surgery, radiotherapy, and chemotherapy provide limited survival benefits due to the tumor's heterogeneity and resistance mechanisms. This review examines novel approaches for treating GBM, focusing on repurposing existing medications such as antipsychotics, antidepressants, and statins for their potential anti-GBM effects. Advances in molecular profiling, including next-generation sequencing, artificial intelligence (AI), and nanotechnology-based drug delivery, are transforming GBM diagnosis and treatment. The TME, particularly GBM stem cells and immune evasion, plays a key role in therapeutic resistance. Integrating multi-omics data and applying precision medicine show promise, especially in combination therapies and immunotherapies, to enhance clinical outcomes. Addressing challenges such as drug resistance, targeting GBM stem cells, and crossing the blood-brain barrier is essential for improving treatment efficacy. While current treatments offer limited benefits, emerging strategies such as immunotherapies, precision medicine, and drug repurposing show significant potential. Technologies like liquid biopsies, AI-powered diagnostics, and nanotechnology could help overcome obstacles like the blood-brain barrier and GBM stem cells. Ongoing research into combination therapies, targeted drug delivery, and personalized treatments is crucial. Collaborative efforts and robust clinical trials are necessary to translate these innovations into effective therapies, offering hope for improved survival and quality of life for GBM patients.
Collapse
Affiliation(s)
- Mohamed S Anwer
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Mohammed A Abdel-Rasol
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt.
| | - Wael M El-Sayed
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt.
| |
Collapse
|
5
|
Pouyan A, Ghorbanlo M, Eslami M, Jahanshahi M, Ziaei E, Salami A, Mokhtari K, Shahpasand K, Farahani N, Meybodi TE, Entezari M, Taheriazam A, Hushmandi K, Hashemi M. Glioblastoma multiforme: insights into pathogenesis, key signaling pathways, and therapeutic strategies. Mol Cancer 2025; 24:58. [PMID: 40011944 PMCID: PMC11863469 DOI: 10.1186/s12943-025-02267-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/07/2025] [Indexed: 02/28/2025] Open
Abstract
Glioblastoma multiforme (GBM) is the most prevalent and aggressive primary brain tumor in adults, characterized by a poor prognosis and significant resistance to existing treatments. Despite progress in therapeutic strategies, the median overall survival remains approximately 15 months. A hallmark of GBM is its intricate molecular profile, driven by disruptions in multiple signaling pathways, including PI3K/AKT/mTOR, Wnt, NF-κB, and TGF-β, critical to tumor growth, invasion, and treatment resistance. This review examines the epidemiology, molecular mechanisms, and therapeutic prospects of targeting these pathways in GBM, highlighting recent insights into pathway interactions and discovering new therapeutic targets to improve patient outcomes.
Collapse
Affiliation(s)
- Ashkan Pouyan
- Department of Neurosurgery, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Masoud Ghorbanlo
- Department of Anesthesiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Eslami
- Department of Neurosurgery, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Jahanshahi
- Department of Neurosurgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Ziaei
- Department of Neurosurgery, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Salami
- Department of Neurosurgery, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khatere Mokhtari
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Koorosh Shahpasand
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Laboratory Medicine and Pathology, Institute for Translational Neuroscience, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Tohid Emami Meybodi
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Functional Neurosurgery Research Center, Shohada Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Epidemiology, University of Tehran, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
6
|
Wong T, Kang R, Yun K. The multi-faceted immune modulatory role of S100A4 in cancer and chronic inflammatory disease. Front Immunol 2025; 16:1525567. [PMID: 40078995 PMCID: PMC11897520 DOI: 10.3389/fimmu.2025.1525567] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
S100A4 is a Ca2+-binding protein involved in multiple chronic inflammatory and neoplastic conditions. This review focuses on recent advances in the understanding of S100A4 function in immune cells, comparing and contrasting S100A4 regulation of immune responses in cancer and chronic inflammatory diseases. We provide evidence that S100A4 regulation of immune cell function has a profound role in promoting the pathogenesis of cancer and pro-inflammatory conditions. Finally, we discuss relevant future directions to target S100A4 therapeutically in different disease states.
Collapse
Affiliation(s)
- Thomas Wong
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, United States
- College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Reece Kang
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, United States
| | - Kyuson Yun
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, United States
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
7
|
Rodriguez-Iglesias N, Paris I, Valero J, Cañas-Zabala L, Carretero A, Hatje K, Zhang JD, Patsch C, Britschgi M, Gutbier S, Sierra A. A bottom-up approach identifies the antipsychotic and antineoplastic trifluoperazine and the ribose derivative deoxytubercidin as novel microglial phagocytosis inhibitors. Glia 2025; 73:330-351. [PMID: 39495090 DOI: 10.1002/glia.24637] [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: 06/04/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024]
Abstract
Phagocytosis is an indispensable function of microglia, the brain professional phagocytes. Microglia is particularly efficient phagocytosing cells that undergo programmed cell death (apoptosis) in physiological conditions. However, mounting evidence suggests microglial phagocytosis dysfunction in multiple brain disorders. These observations prompted us to search for phagocytosis modulators (enhancers or inhibitors) with therapeutic potential. We used a bottom-up strategy that consisted on the identification of phagocytosis modulators using phenotypic high throughput screenings (HTSs) in cell culture and validation in organotypic cultures and in vivo. We performed two complementary HTS campagnes: at Achucarro, we used primary cultures of mouse microglia and compounds of the Prestwick Chemical Library; at Roche, we used human iPSC derived macrophage-like cells and a proprietary chemo-genomic library with 2200 compounds with known mechanism-of-action. Next, we validated the more robust compounds using hippocampal organotypic cultures and identified two phagocytosis inhibitors: trifluoperazine, a dopaminergic and adrenergic antagonist used as an antipsychotic and antineoplastic; and deoxytubercidin, a ribose derivative. Finally, we tested whether these compounds were able to modulate phagocytosis of apoptotic newborn cells in the adult hippocampal neurogenic niche in vivo by administering them into the mouse hippocampus using osmotic minipumps. We confirmed that both trifluoperazine and deoxytubercidin have anti-phagocytic activity in vivo, and validated our bottom-up strategy to identify novel phagocytosis modulators. These results show that chemical libraries with annotated mechanism of action are an starting point for the pharmacological modulation of microglia in drug discovery projects aiming at the therapeutic manipulation of phagocytosis in brain diseases.
Collapse
Affiliation(s)
- Noelia Rodriguez-Iglesias
- Glial Cell Biology Lab, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neuroscience, University of the Basque Country EHU/UPV, Leioa, Spain
| | - Iñaki Paris
- Glial Cell Biology Lab, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neuroscience, University of the Basque Country EHU/UPV, Leioa, Spain
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jorge Valero
- Glial Cell Biology Lab, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neuroscience, University of the Basque Country EHU/UPV, Leioa, Spain
- Ikerbasque Foundation, Bilbao, Spain
| | - Lorena Cañas-Zabala
- Glial Cell Biology Lab, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neuroscience, University of the Basque Country EHU/UPV, Leioa, Spain
| | - Alejandro Carretero
- Glial Cell Biology Lab, Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Klas Hatje
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jitao David Zhang
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Christoph Patsch
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Markus Britschgi
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Simon Gutbier
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Amanda Sierra
- Glial Cell Biology Lab, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque Foundation, Bilbao, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country EHU/UPV, Leioa, Spain
| |
Collapse
|
8
|
Sun S, Shyr Z, McDaniel K, Fang Y, Tao D, Chen CZ, Zheng W, Zhu Q. Reversal gene expression assessment for drug repurposing, a case study of glioblastoma. J Transl Med 2025; 23:25. [PMID: 39773231 PMCID: PMC11706105 DOI: 10.1186/s12967-024-06046-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/25/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a rare brain cancer with an exceptionally high mortality rate, which illustrates the pressing demand for more effective therapeutic options. Despite considerable research efforts on GBM, its underlying biological mechanisms remain unclear. Furthermore, none of the United States Food and Drug Administration (FDA) approved drugs used for GBM deliver satisfactory survival improvement. METHODS This study presents a novel computational pipeline by utilizing gene expression data analysis for GBM for drug repurposing to address the challenges in rare disease drug development, particularly focusing on GBM. The GBM Gene Expression Profile (GGEP) was constructed with multi-omics data to identify drugs with reversal gene expression to GGEP from the Integrated Network-Based Cellular Signatures (iLINCS) database. RESULTS We prioritized the candidates via hierarchical clustering of their expression signatures and quantification of their reversal strength by calculating two self-defined indices based on the GGEP genes' log2 foldchange (LFC) that the drug candidates could induce. Among five prioritized candidates, in-vitro experiments validated Clofarabine and Ciclopirox as highly efficacious in selectively targeting GBM cancer cells. CONCLUSIONS The success of this study illustrated a promising avenue for accelerating drug development by uncovering underlying gene expression effect between drugs and diseases, which can be extended to other rare diseases and non-rare diseases.
Collapse
Affiliation(s)
- Shixue Sun
- Informatics Core, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Zeenat Shyr
- Early Translation Branch, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Kathleen McDaniel
- Early Translation Branch, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Yuhong Fang
- Analytical Chemistry Core, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Dingyin Tao
- Analytical Chemistry Core, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Catherine Z Chen
- Early Translation Branch, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Wei Zheng
- Early Translation Branch, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Qian Zhu
- Informatics Core, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA.
| |
Collapse
|
9
|
Wang H, Cheng C, Ding J, Qian R, Luo T, Zheng L, Chen Y. Trifluoperazine effect on human sperm: The accumulation of reactive oxygen species and the decrease in the mitochondrial membrane potential. Reprod Toxicol 2024; 130:108730. [PMID: 39369966 DOI: 10.1016/j.reprotox.2024.108730] [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: 07/23/2024] [Revised: 09/05/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
A strong link between antipsychotic drug use and reduced human sperm quality has been reported. Trifluoperazine (TFP), a commonly used antipsychotic, is now being explored for anticancer applications. Although there are hints that TFP might affect the male reproductive system, its impact on human sperm quality remains uncertain. Using a human sperm and TFP in vitro coculture system, we examined the effect of TFP (12.5, 25, 50 and 100 μM) on human sperm function and physiological parameters. The results showed that 50 μM and 100 μM TFP induced the accumulation of reactive oxygen species (ROS) and a decrease in the mitochondrial membrane potential (MMP) of human sperm, leading to decreased sperm viability, while 25 μM TFP inhibited only the penetration ability, total sperm motility, and progressive motility. Although 12.5 μM and 25 μM TFP increased [Ca2+]i in human sperm, they did not affect capacitation or the acrosome reaction. These results may be explained by the observation that 12.5 μM and 25 μM TFP did not increase tyrosine phosphorylation in human sperm, although TFP increased [Ca2+]i in a time-course traces similar to that of progesterone. Our results indicated that TFP could cause male reproductive toxicity by inducing the accumulation of ROS and a decrease in the MMP in human sperm.
Collapse
Affiliation(s)
- Houpeng Wang
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; School of Public Health, Jiangxi Medical College, Nanchang University, China
| | - Cheng Cheng
- Institute of Biomedical Innovation and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Jing Ding
- Institute of Biomedical Innovation and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Ruirui Qian
- Institute of Biomedical Innovation and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Tao Luo
- Institute of Biomedical Innovation and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Liping Zheng
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; School of Public Health, Jiangxi Medical College, Nanchang University, China.
| | - Ying Chen
- Institute of Biomedical Innovation and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China.
| |
Collapse
|
10
|
Gharzia FG, Aljohmani A, Beck A, Philipp SE, Yildiz D. Regulation of ADAM10 activity through microdomain-dependent intracellular calcium changes. Cell Commun Signal 2024; 22:531. [PMID: 39497138 PMCID: PMC11533308 DOI: 10.1186/s12964-024-01891-5] [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: 06/11/2024] [Accepted: 10/12/2024] [Indexed: 11/06/2024] Open
Abstract
A disintegrin and metalloproteinases (ADAMs) are transmembrane proteases that cleave other proteins close to the surface in a process called shedding. The prominent member ADAM10 has been linked to several pathologies such as Alzheimer's disease, bacterial infection, cancer development and metastasis. Although the regulation of the ADAM10 activity by calcium influx and calmodulin inhibition has been reported, the spatiotemporal regulation of Ca2+-dependent ADAM10 activation and the required source of Ca2+ ions have not been thoroughly studied. In the present study, we observed the rapid Ca2+-dependent activation of ADAM10 in A549 lung carcinoma cells upon stimulation with ionomycin. The calmodulin-inhibitors trifluoperazine and ophiobolin A mediated delayed activation of ADAM10, which apparently did not depend on intracellular Ca2+ in the case of trifluoperazine. Furthermore, the surface translocation and release of ADAM10 in extracellular vesicles exhibited different kinetics and were only partially linked to catalytic activation. Finally, ADAM10 activation was observed after the entry of Ca2+ through certain channels, such as canonical members of transient receptor potential (TRP) channels. Therefore, the opening of particular channels for Ca2+ entry points and subsequent Ca2+ flux as well as the temporal aspects of the consequent increase in Ca2+ levels, must be considered for future therapeutic options involving the increasing or decreasing ADAM10 activity.
Collapse
Affiliation(s)
| | - Ahmad Aljohmani
- Molecular Pharmacology, PZMS, Saarland University, Campus Homburg Building 46, 66421, Homburg, Germany
| | - Andreas Beck
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, Homburg, Germany
| | - Stephan E Philipp
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, Homburg, Germany
| | - Daniela Yildiz
- Molecular Pharmacology, PZMS, Saarland University, Campus Homburg Building 46, 66421, Homburg, Germany.
| |
Collapse
|
11
|
Dai M, Chen Y, Qin J. Atrioventricular re-entrant tachycardia and atrioventricular node re-entrant tachycardia in a patient with cancer under chemotherapy: a case report and literature review. Front Cardiovasc Med 2024; 11:1367893. [PMID: 38911514 PMCID: PMC11190324 DOI: 10.3389/fcvm.2024.1367893] [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: 01/09/2024] [Accepted: 05/21/2024] [Indexed: 06/25/2024] Open
Abstract
Cardio-oncology is a new field of interest in cardiology focusing on the detection and treatment of cardiovascular diseases, such as arrhythmias, myocarditis, and heart failure, as side-effects of chemotherapy and radiotherapy. The association between chemotherapeutic agents and arrhythmias has previously been established. Atrial tachyarrhythmias, particularly atrial fibrillation, are most common, but ventricular arrhythmias, including those related to treatment-induced QT prolongation, and bradyarrhythmias can also occur. However, the association between chemotherapeutic agents and atrioventricular re-entrant tachycardia (AVRT)/atrioventricular node re-entrant tachycardia (AVNRT) remains poorly understood. Here, we report a patient with new-onset AVRT/AVNRT and lung cancer who underwent chemotherapy. We considered that chemotherapy or cancer itself may have been a trigger for the initiation of paroxysmal AVRT/AVNRT, and that radiofrequency catheter ablation was effective in treating this type of tachycardia. Here, possible mechanisms and potential genes (mostly ion channels) involved in AVRT/AVNRT are summarized and the mechanisms underlying the possible regulatory patterns of cancer cells and chemotherapy on ion channels are reviewed. Finally, we considered that ion channel abnormalities may link cancer or chemotherapy to the onset of AVRT/AVNRT. The aim of the present study was to highlight the association between chemotherapeutic agents and AVRT/AVNRT and to provide new insights for future research. Understanding the intermediate mechanisms between chemotherapeutic agents and AVRT/AVNRT may be beneficial in preventing chemotherapy-evoked AVRT/AVNRT (and/or other arrhythmias) in future.
Collapse
Affiliation(s)
- Meiyan Dai
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Chen
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin Qin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Yang J, Li S, Wang J, Liu G, Zhang C, Li X, Liu X. Calmodulin 2 expression is associated with poor prognosis in breast cancer. Pathol Res Pract 2024; 258:155326. [PMID: 38754328 DOI: 10.1016/j.prp.2024.155326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/13/2024] [Accepted: 04/21/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Calmodulin 2 (CALM2) belongs to the highly conserved calcium-binding protein family, implicated in the pathogenesis of various malignant tumors. However, its involvement in breast cancer (BRCA) remains unclear. This study aimed to examine CALM2 expression in BRCA and its associations with prognosis, clinicopathological features, protein-protein interactions, and immune cell infiltration. MATERIALS AND METHODS Online bioinformatics tools were employed to assess CALM2 expression and its clinical relevance in BRCA. Western blotting and immunohistochemistry were utilized to evaluate CALM2 expression in BRCA cell lines and tissues. Logistic regression was applied to analyze the relationship between CALM2 expression levels and clinicopathological parameters. Transwell assay was performed to validate the role of CALM2 in BRCA migration and invasion. RESULTS CALM2 expression was significantly elevated in BRCA, with increased levels predicting poor overall survival (OS) and disease-free survival (DFS). Moreover, high CALM2 expression correlated with poorer DFS specifically in triple-negative breast cancer (TNBC). CALM2 expression in BRCA showed significant associations with lymph node metastasis, TP53 mutation status, and menopause status. Silencing CALM2 in BRCA cells demonstrated inhibition of cell migration and invasion in vitro. CONCLUSIONS CALM2 is overexpressed in BRCA and its upregulation is significantly correlated with poor patient prognosis. Elevated CALM2 expression holds promise as a potential molecular marker for predicting poor survival and as a therapeutic target in BRCA.
Collapse
Affiliation(s)
- Ju Yang
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Shuixian Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jigang Wang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guoyuan Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chenyang Zhang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaojing Li
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Xiuping Liu
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China; Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
13
|
Mubeen S, Raza I, Ujjan B, Wasim B, Khan L, Naeem N, Enam SA, Hanif F. Iloperidone and Temozolomide Synergistically Inhibit Growth, Migration and Enhance Apoptosis in Glioblastoma Cells. Biomedicines 2024; 12:1134. [PMID: 38927341 PMCID: PMC11200733 DOI: 10.3390/biomedicines12061134] [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: 04/09/2024] [Revised: 05/06/2024] [Accepted: 05/14/2024] [Indexed: 06/28/2024] Open
Abstract
Glioblastoma (GBM) is a fatal astrocytic glioma with poor prognosis and treatment resistance. Repurposing potential FDA-approved drugs like anti-psychotics can address the concerns in a timely and cost-effective manner. Epidemiological studies have shown that patients with schizophrenic using anti-psychotics have a low incidence of GBM. Therefore, we aimed to investigate the therapeutic potential of atypical anti-psychotic Iloperidone (ILO) alone and in combination with Temozolomide (TMZ) against GBM. The study assessed the growth inhibitory effect of ILO, TMZ, and their combination (ILO + TMZ) on U-87MG and T-98G cell lines using an MTT assay. The drug interaction coefficient (CDI) was determined, and doses with synergistic effects were used for subsequent experiments, including migratory, invasion, and TUNEL assays. The expressions of DRD2, β-catenin, Dvl2, Twist, and Slug were assessed by RTq-PCR, whereas the β-catenin protein expression was also determined by immunocytochemistry. ILO (p < 0.05) and TMZ (p < 0.01) significantly inhibited the growth of U-87MG cells at all tested doses. The combination of 60 µM of both drugs showed synergistic activity with CDI < 1. The inhibition of migration and apoptosis was more pronounced in the case of combination treatment (p < 0.001). Inhibition of the invading cells was also found to be significant in ILO- and combination-treated groups (p < 0.001). ILO and combination treatment also significantly downregulated the expression of DRD2, while TMZ upregulated the expression (p < 0.001). The expressions of β-catenin (p < 0.001), Dvl2 (p < 0.001), Twist (p < 0.001), and Slug (p < 0.001) were also significantly downregulated in all treatment groups as compared to the vehicle control. The data suggest that ILO possesses strong growth inhibitory activity, possibly due to its effect on DRD2 and β-catenin expression and has the potential to be repurposed against GBM.
Collapse
Affiliation(s)
- Sahar Mubeen
- Department of Anatomy, Dow International Medical College, Dow University of Health Sciences, Karachi 75330, Pakistan;
| | - Iffat Raza
- Department of Anatomy, Karachi Institute of Medical Sciences, Karachi 75080, Pakistan;
| | - Badaruddin Ujjan
- Department of Neurosurgery, Dow University Hospital, Dow University of Health Sciences, Karachi 74200, Pakistan;
| | - Bushra Wasim
- Department of Anatomy, Ziauddin University Hospital, Karachi 75600, Pakistan;
| | - Lubna Khan
- Department of Biochemistry, Dow International Medical College, Dow University of Health Sciences, Karachi 75330, Pakistan;
| | - Nadia Naeem
- Dow Research Institute of Biotechnology & Biomedical Sciences, Karachi 75330, Pakistan;
| | - Syed Ather Enam
- Center of Oncological Research in Surgery, Aga Khan University Hospital, Karachi 74800, Pakistan;
| | - Farina Hanif
- Department of Biochemistry, Dow International Medical College, Dow University of Health Sciences, Karachi 75330, Pakistan;
| |
Collapse
|
14
|
Wang J, Choi WG, Nguyen NK, Liu D, Kim SH, Lim D, Hwang BK, Jwa NS. Cytoplasmic Ca 2+ influx mediates iron- and reactive oxygen species-dependent ferroptotic cell death in rice immunity. FRONTIERS IN PLANT SCIENCE 2024; 15:1339559. [PMID: 38756966 PMCID: PMC11096502 DOI: 10.3389/fpls.2024.1339559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/17/2024] [Indexed: 05/18/2024]
Abstract
Iron- and reactive oxygen species (ROS)-dependent ferroptosis occurs in plant cells. Ca2+ acts as a conserved key mediator to control plant immune responses. Here, we report a novel role of cytoplasmic Ca2+ influx regulating ferroptotic cell death in rice immunity using pharmacological approaches. High Ca2+ influx triggered iron-dependent ROS accumulation, lipid peroxidation, and subsequent hypersensitive response (HR) cell death in rice (Oryza sativa). During Magnaporthe oryzae infection, 14 different Ca2+ influx regulators altered Ca2+, ROS and Fe2+ accumulation, glutathione reductase (GR) expression, glutathione (GSH) depletion and lipid peroxidation, leading to ferroptotic cell death in rice. High Ca2+ levels inhibited the reduction of glutathione isulphide (GSSG) to GSH in vitro. Ca2+ chelation by ethylene glycol-bis (2-aminoethylether)-N, N, N', N'-tetra-acetic acid (EGTA) suppressed apoplastic Ca2+ influx in rice leaf sheaths during infection. Blocking apoplastic Ca2+ influx into the cytoplasm by Ca2+ chelation effectively suppressed Ca2+-mediated iron-dependent ROS accumulation and ferroptotic cell death. By contrast, acibenzolar-S-methyl (ASM), a plant defense activator, significantly enhanced Ca2+ influx, as well as ROS and iron accumulation to trigger ferroptotic cell death in rice. The cytoplasmic Ca2+ influx through calcium-permeable cation channels, including the putative resistosomes, could mediate iron- and ROS-dependent ferroptotic cell death under reduced GR expression levels in rice immune responses.
Collapse
Affiliation(s)
- Juan Wang
- Division of Integrative Bioscience and Biotechnology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Won-Gyu Choi
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, United States
| | - Nam Khoa Nguyen
- Division of Integrative Bioscience and Biotechnology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Dongping Liu
- Division of Integrative Bioscience and Biotechnology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Su-Hwa Kim
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, United States
| | - Dongyeol Lim
- Department of Chemistry, College of Natural Sciences, Sejong University, Seoul, Republic of Korea
| | - Byung Kook Hwang
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Nam-Soo Jwa
- Division of Integrative Bioscience and Biotechnology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| |
Collapse
|
15
|
Sun X, Wu L, Du L, Xu W, Han M. Targeting the organelle for radiosensitization in cancer radiotherapy. Asian J Pharm Sci 2024; 19:100903. [PMID: 38590796 PMCID: PMC10999375 DOI: 10.1016/j.ajps.2024.100903] [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: 10/06/2023] [Revised: 12/29/2023] [Accepted: 01/16/2024] [Indexed: 04/10/2024] Open
Abstract
Radiotherapy is a well-established cytotoxic therapy for local solid cancers, utilizing high-energy ionizing radiation to destroy cancer cells. However, this method has several limitations, including low radiation energy deposition, severe damage to surrounding normal cells, and high tumor resistance to radiation. Among various radiotherapy methods, boron neutron capture therapy (BNCT) has emerged as a principal approach to improve the therapeutic ratio of malignancies and reduce lethality to surrounding normal tissue, but it remains deficient in terms of insufficient boron accumulation as well as short retention time, which limits the curative effect. Recently, a series of radiosensitizers that can selectively accumulate in specific organelles of cancer cells have been developed to precisely target radiotherapy, thereby reducing side effects of normal tissue damage, overcoming radioresistance, and improving radiosensitivity. In this review, we mainly focus on the field of nanomedicine-based cancer radiotherapy and discuss the organelle-targeted radiosensitizers, specifically including nucleus, mitochondria, endoplasmic reticulum and lysosomes. Furthermore, the organelle-targeted boron carriers used in BNCT are particularly presented. Through demonstrating recent developments in organelle-targeted radiosensitization, we hope to provide insight into the design of organelle-targeted radiosensitizers for clinical cancer treatment.
Collapse
Affiliation(s)
- Xiaoyan Sun
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Linjie Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Lina Du
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Wenhong Xu
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Afliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Min Han
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Afliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
16
|
Zhang W, Chen S, Ma B, Ding Y, Liu X, He C, Wang B, Yuan M. Trifluoperazine regulates blood-brain barrier permeability via the MLCK/p-MLC pathway to promote ischemic stroke recovery. iScience 2024; 27:109156. [PMID: 38439960 PMCID: PMC10910233 DOI: 10.1016/j.isci.2024.109156] [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: 09/18/2023] [Revised: 12/19/2023] [Accepted: 02/02/2024] [Indexed: 03/06/2024] Open
Abstract
Blood-brain barrier (BBB) disruption following ischemic stroke (IS) can induce significant aftereffects. Elevated calmodulin (CaM) expression following stroke causes calcium overload-a key contributor to BBB collapse. Trifluoperazine (TFP), a CaM inhibitor, reduces CaM overexpression following IS. However, it remains unclear whether TFP participates in BBB repair after IS. We administered TFP to mice subjected to middle cerebral artery occlusion (MCAO) and bEnd.3 cells subjected to oxygen-glucose deprivation (OGD). TFP treatment in MCAO mice reduced cerebral CaM expression and infarct size and decreased BBB permeability. OGD-treated bEnd.3 cells showed significantly increased CaM protein levels and reduced tight junction (TJ) protein levels; these changes were reversed by TFP treatment. Our results found that TFP administration in mice inhibited actin contraction following cerebral ischemia-reperfusion by suppressing the MLCK/p-MLC pathway, thereby attenuating cell retraction, improving TJ protein integrity, and reducing BBB permeability. Consequently, this treatment may promote neurological function recovery after IS.
Collapse
Affiliation(s)
- Wentao Zhang
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Sisi Chen
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Bin Ma
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yingmei Ding
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiaofen Liu
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Caijun He
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Biao Wang
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Mei Yuan
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Affiliated Nanhua Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
17
|
Onciul R, Brehar FM, Toader C, Covache-Busuioc RA, Glavan LA, Bratu BG, Costin HP, Dumitrascu DI, Serban M, Ciurea AV. Deciphering Glioblastoma: Fundamental and Novel Insights into the Biology and Therapeutic Strategies of Gliomas. Curr Issues Mol Biol 2024; 46:2402-2443. [PMID: 38534769 DOI: 10.3390/cimb46030153] [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: 01/24/2024] [Revised: 03/06/2024] [Accepted: 03/09/2024] [Indexed: 03/28/2024] Open
Abstract
Gliomas constitute a diverse and complex array of tumors within the central nervous system (CNS), characterized by a wide range of prognostic outcomes and responses to therapeutic interventions. This literature review endeavors to conduct a thorough investigation of gliomas, with a particular emphasis on glioblastoma (GBM), beginning with their classification and epidemiological characteristics, evaluating their relative importance within the CNS tumor spectrum. We examine the immunological context of gliomas, unveiling the intricate immune environment and its ramifications for disease progression and therapeutic strategies. Moreover, we accentuate critical developments in understanding tumor behavior, focusing on recent research breakthroughs in treatment responses and the elucidation of cellular signaling pathways. Analyzing the most novel transcriptomic studies, we investigate the variations in gene expression patterns in glioma cells, assessing the prognostic and therapeutic implications of these genetic alterations. Furthermore, the role of epigenetic modifications in the pathogenesis of gliomas is underscored, suggesting that such changes are fundamental to tumor evolution and possible therapeutic advancements. In the end, this comparative oncological analysis situates GBM within the wider context of neoplasms, delineating both distinct and shared characteristics with other types of tumors.
Collapse
Affiliation(s)
- Razvan Onciul
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Neurosurgery Department, Emergency University Hospital, 050098 Bucharest, Romania
| | - Felix-Mircea Brehar
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Neurosurgery, Clinical Emergency Hospital "Bagdasar-Arseni", 041915 Bucharest, Romania
| | - Corneliu Toader
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | | | - Luca-Andrei Glavan
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Bogdan-Gabriel Bratu
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Horia Petre Costin
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - David-Ioan Dumitrascu
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Matei Serban
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
| |
Collapse
|
18
|
Song Y, Xue T, Guo S, Yu Z, Yun C, Zhao J, Song Z, Liu Z. Inhibition of aquaporin-4 and its subcellular localization attenuates below-level central neuropathic pain by regulating astrocyte activation in a rat spinal cord injury model. Neurotherapeutics 2024; 21:e00306. [PMID: 38237380 DOI: 10.1016/j.neurot.2023.e00306] [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: 11/19/2023] [Indexed: 03/24/2024] Open
Abstract
The mechanisms of central neuropathic pain (CNP) caused by spinal cord injury have not been sufficiently studied. We have found that the upregulation of astrocytic aquaporin-4 (AQP4) aggravated peripheral neuropathic pain after spinal nerve ligation in rats. Using a T13 spinal cord hemisection model, we showed that spinal AQP4 was markedly upregulated after SCI and mainly expressed in astrocytes in the spinal dorsal horn (SDH). Inhibition of AQP4 with TGN020 suppressed astrocyte activation, attenuated the development and maintenance of below-level CNP and promoted motor function recovery in vivo. In primary astrocyte cultures, TGN020 also changed cell morphology, diminished cell proliferation and suppressed astrocyte activation. Moreover, T13 spinal cord hemisection induced cell-surface abundance of the AQP4 channel and perivascular localization in the SDH. Targeted inhibition of AQP4 subcellular localization with trifluoperazine effectively diminished astrocyte activation in vitro and further ablated astrocyte activation, attenuated the development and maintenance of below-level CNP, and accelerated functional recovery in vivo. Together, these results provide mechanistic insights into the roles of AQP4 in the development and maintenance of below-level CNP. Intervening with AQP4, including targeting AQP4 subcellular localization, might emerge as a promising agent to prevent chronic CNP after SCI.
Collapse
Affiliation(s)
- Yu Song
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Tao Xue
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, China
| | - Shiwu Guo
- Department of Orthopedics, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, 215028, China
| | - Zhen Yu
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Chengming Yun
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, China
| | - Jie Zhao
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, China
| | - Zhiwen Song
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Zhiyuan Liu
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, China; Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China; The Wujin Clinical College of Xuzhou Medical University, Changzhou 213003, China.
| |
Collapse
|
19
|
Bond ACS, Crocker MA, Wilczek MP, DuShane JK, Sandberg AL, Bennett LJ, Leclerc NR, Maginnis MS. High-throughput drug screen identifies calcium and calmodulin inhibitors that reduce JCPyV infection. Antiviral Res 2024; 222:105817. [PMID: 38246207 PMCID: PMC10922812 DOI: 10.1016/j.antiviral.2024.105817] [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: 11/18/2023] [Revised: 01/09/2024] [Accepted: 01/18/2024] [Indexed: 01/23/2024]
Abstract
JC polyomavirus (JCPyV) is a nonenveloped, double-stranded DNA virus that infects the majority of the population. Immunocompetent individuals harbor infection in their kidneys, while severe immunosuppression can result in JCPyV spread to the brain, causing the neurodegenerative disease progressive multifocal leukoencephalopathy (PML). Due to a lack of approved therapies to treat JCPyV and PML, the disease results in rapid deterioration, and is often fatal. In order to identify potential antiviral treatments for JCPyV, a high-throughput, large-scale drug screen was performed using the National Institutes of Health Clinical Collection (NCC). Drugs from the NCC were tested for inhibitory effects on JCPyV infection, and drugs from various classes that reduced JCPyV infection were identified, including receptor agonists and antagonists, calcium signaling modulators, and enzyme inhibitors. Given the role of calcium signaling in viral infection including Merkel cell polyomavirus and simian virus 40 polyomavirus (SV40), calcium signaling inhibitors were further explored for the capacity to impact JCPyV infection. Calcium and calmodulin inhibitors trifluoperazine (TFP), W-7, tetrandrine, and nifedipine reduced JCPyV infection, and TFP specifically reduced viral internalization. Additionally, TFP and W-7 reduced infection by BK polyomavirus, SV40, and SARS-CoV-2. These results highlight specific inhibitors, some FDA-approved, for the possible treatment and prevention of JCPyV and several other viruses, and further illuminate the calcium and calmodulin pathway as a potential target for antiviral drug development.
Collapse
Affiliation(s)
- Avery C S Bond
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME, 04469, USA
| | - Mason A Crocker
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME, 04469, USA
| | - Michael P Wilczek
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME, 04469, USA
| | - Jeanne K DuShane
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME, 04469, USA
| | - Amanda L Sandberg
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME, 04469, USA
| | - Lucas J Bennett
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME, 04469, USA
| | - Nicholas R Leclerc
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME, 04469, USA
| | - Melissa S Maginnis
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME, 04469, USA; Graduate School of Biomedical Science and Engineering, Orono, ME, 04469, USA.
| |
Collapse
|
20
|
Kreitzer MA, Vredeveld M, Tinner K, Powell AM, Schantz AW, Leininger R, Merillat R, Gongwer MW, Tchernookova BK, Malchow RP. ATP-mediated increase in H + efflux from retinal Müller cells of the axolotl. J Neurophysiol 2024; 131:124-136. [PMID: 38116604 PMCID: PMC11286307 DOI: 10.1152/jn.00321.2023] [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: 08/25/2023] [Revised: 11/17/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023] Open
Abstract
Previous work has shown that activation of tiger salamander retinal radial glial cells by extracellular ATP induces a pronounced extracellular acidification, which has been proposed to be a potent modulator of neurotransmitter release. This study demonstrates that low micromolar concentrations of extracellular ATP similarly induce significant H+ effluxes from Müller cells isolated from the axolotl retina. Müller cells were enzymatically isolated from axolotl retina and H+ fluxes were measured from individual cells using self-referencing H+-selective microelectrodes. The increased H+ efflux from axolotl Müller cells induced by extracellular ATP required activation of metabotropic purinergic receptors and was dependent upon calcium released from internal stores. We further found that the ATP-evoked increase in H+ efflux from Müller cells of both tiger salamander and axolotl were sensitive to pharmacological agents known to interrupt calmodulin and protein kinase C (PKC) activity: chlorpromazine (CLP), trifluoperazine (TFP), and W-7 (all calmodulin inhibitors) and chelerythrine, a PKC inhibitor, all attenuated ATP-elicited increases in H+ efflux. ATP-initiated H+ fluxes of axolotl Müller cells were also significantly reduced by amiloride, suggesting a significant contribution by sodium-hydrogen exchangers (NHEs). In addition, α-cyano-4-hydroxycinnamate (4-cin), a monocarboxylate transport (MCT) inhibitor, also reduced the ATP-induced increase in H+ efflux in both axolotl and tiger salamander Müller cells, and when combined with amiloride, abolished ATP-evoked increase in H+ efflux. These data suggest that axolotl Müller cells are likely to be an excellent model system to understand the cell-signaling pathways regulating H+ release from glia and the role this may play in modulating neuronal signaling.NEW & NOTEWORTHY Glial cells are a key structural part of the tripartite synapse and have been suggested to regulate synaptic transmission, but the regulatory mechanisms remain unclear. We show that extracellular ATP, a potent glial cell activator, induces H+ efflux from axolotl retinal Müller (glial) cells through a calcium-dependent pathway that is likely to involve calmodulin, PKC, Na+/H+ exchange, and monocarboxylate transport, and suggest that such H+ release may play a key role in modulating neuronal transmission.
Collapse
Affiliation(s)
- Matthew A Kreitzer
- Department of Biology, Indiana Wesleyan University, Marion, Indiana, United States
| | - Mason Vredeveld
- Department of Biology, Indiana Wesleyan University, Marion, Indiana, United States
| | - Kaleb Tinner
- Department of Biology, Indiana Wesleyan University, Marion, Indiana, United States
| | - Alyssa M Powell
- Department of Biology, Indiana Wesleyan University, Marion, Indiana, United States
| | - Adam W Schantz
- Department of Biology, Indiana Wesleyan University, Marion, Indiana, United States
| | - Rachel Leininger
- Department of Biology, Indiana Wesleyan University, Marion, Indiana, United States
| | - Rajapone Merillat
- Department of Biology, Indiana Wesleyan University, Marion, Indiana, United States
| | - Michael W Gongwer
- Department of Biology, Indiana Wesleyan University, Marion, Indiana, United States
| | - Boriana K Tchernookova
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Robert Paul Malchow
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Psychology, College of the Holy Cross, Worcester, Massachusetts, United States
| |
Collapse
|
21
|
Sammeta SS, Banarase TA, Rahangdale SR, Wankhede NL, Aglawe MM, Taksande BG, Mangrulkar SV, Upaganlawar AB, Koppula S, Kopalli SR, Umekar MJ, Kale MB. Molecular understanding of ER-MT communication dysfunction during neurodegeneration. Mitochondrion 2023; 72:59-71. [PMID: 37495165 DOI: 10.1016/j.mito.2023.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/13/2023] [Accepted: 07/23/2023] [Indexed: 07/28/2023]
Abstract
Biological researchers are seeing organelles in a new light. These cellular entities have been believed to be singular and distinctive structures that performed specialized purposes for a very long time. But in recentpast years, scientists have learned that organelles become dynamic and make physical contact. Additionally, Biological processes are regulated by organelles interactions and its alteration play an important role in cell malfunctioning and several pathologies, including neurodegenerative diseases. Mitochondrial-ER contact sites (MERCS) have received considerable attention in the domain of cell homeostasis and dysfunction, specifically in the area of neurodegeneration. This is largely due to the significant role of this subcellular compartment in a diverse array of vital cellular functions, including Ca2+ homeostasis, transport, bioenergetics and turnover, mitochondrial dynamics, apoptotic signaling, ER stress, and inflammation. A significant number of disease-associated proteins were found to physically interact with the ER-Mitochondria (ER-MT) interface, causing structural and/or functional alterations in this compartment. In this review, we summarize current knowledge about the structure and functions of the ER-MT contact sites, as well as the possible repercussions of their alteration in notable neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and fronto-temporal dementia. The constraints and complexities in defining the nature and origin of the highlighted defects in ER-MT communication, as well as their concise contribution to the neurodegenerative process, are illustrated in particular. The possibility of using MERCS as a potential drug target to prevent neuronal damage and ultimately neurodegeneration is the topic of our final discussion.
Collapse
Affiliation(s)
- Shivkumar S Sammeta
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Trupti A Banarase
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sandip R Rahangdale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Manish M Aglawe
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Shubhada V Mangrulkar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| |
Collapse
|
22
|
Nafe R, Hattingen E. The Spectrum of Molecular Pathways in Gliomas-An Up-to-Date Review. Biomedicines 2023; 11:2281. [PMID: 37626776 PMCID: PMC10452344 DOI: 10.3390/biomedicines11082281] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
During the last 20 years, molecular alterations have gained increasing significance in the diagnosis and biological assessment of tumors. Gliomas represent the largest group of tumors of the central nervous system, and the main aim of this review is to present the current knowledge on molecular pathways and their alterations in gliomas. A wide range of new insights has been gained, including evidence for the involvement of the WNT pathway or the hippo pathway in the pathobiology of gliomas, indicating a broad involvement of different pathways formerly not considered to play a central role in gliomas. Even new aspects of angiogenic, apoptotic, and metabolic pathways are presented, as well as the rapidly growing field of epigenetic processes, including non-coding RNAs. The two major conclusions drawn from the present review are the distinct interconnectivity of the whole spectrum of molecular pathways and the prominent role of non-coding RNAs, especially circular RNAs, in the regulation of specific targets. All these new insights are discussed, even considering the topic of the resistance to therapy of gliomas, along with aspects that are still incompletely understood, like the role of hydroxymethylation, or even ferroptosis, in the pathobiology of gliomas.
Collapse
Affiliation(s)
- Reinhold Nafe
- Department of Neuroradiology, Clinics of Johann Wolfgang Goethe-University, Schleusenweg 2-16, D-60528 Frankfurt am Main, Germany;
| | | |
Collapse
|
23
|
Yeh CF, Lee WY, Yu TH, Hsu YB, Lan MC, Lan MY. Antipsychotic drug trifluoperazine as a potential therapeutic agent against nasopharyngeal carcinoma. Head Neck 2023; 45:316-328. [PMID: 36349408 DOI: 10.1002/hed.27238] [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: 03/30/2022] [Revised: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Trifluoperazine (TFP) is a typical antipsychotic primarily used to treat schizophrenia. In this study, we aimed to evaluate whether TFP can be used as a therapeutic agent against nasopharyngeal carcinoma (NPC) and identify its underlying molecular mechanisms. METHODS We used NPC-TW01, TW03, TW04, and BM to assess the anticancer effects of TFP by using cytotoxicity, wound healing, colony formation, and cell invasion assays. An in vivo animal study was conducted. RNA sequencing combined with Ingenuity Pathways Analysis was performed to identify the mechanism by which TFP influences NPC cells. RESULTS Our data revealed that TFP decreased NPC cell viability in a dose-dependent manner. The invasion and migration of NPC tumor cells were inhibited by TFP. An in vivo study also demonstrated the anticancer effects of TFP. RNA sequencing revealed several anticancer molecular mechanisms following TFP administration. CONCLUSIONS The antipsychotic drug TFP could be a potential therapeutic regimen for NPC treatment.
Collapse
Affiliation(s)
- Chien-Fu Yeh
- Department of Otorhinolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Otorhinolaryngology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wen-Ya Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Otorhinolaryngology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-Han Yu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yen-Bin Hsu
- Department of Otorhinolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Otorhinolaryngology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Chin Lan
- Department of Otolaryngology-Head and Neck Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Ming-Ying Lan
- Department of Otorhinolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Otorhinolaryngology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
24
|
Jiang T, Wang Q, Lv J, Lin L. Mitochondria-endoplasmic reticulum contacts in sepsis-induced myocardial dysfunction. Front Cell Dev Biol 2022; 10:1036225. [PMID: 36506093 PMCID: PMC9730255 DOI: 10.3389/fcell.2022.1036225] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022] Open
Abstract
Mitochondrial and endoplasmic reticulum (ER) are important intracellular organelles. The sites that mitochondrial and ER are closely related in structure and function are called Mitochondria-ER contacts (MERCs). MERCs are involved in a variety of biological processes, including calcium signaling, lipid synthesis and transport, autophagy, mitochondrial dynamics, ER stress, and inflammation. Sepsis-induced myocardial dysfunction (SIMD) is a vital organ damage caused by sepsis, which is closely associated with mitochondrial and ER dysfunction. Growing evidence strongly supports the role of MERCs in the pathogenesis of SIMD. In this review, we summarize the biological functions of MERCs and the roles of MERCs proteins in SIMD.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiagao Lv
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Jiagao Lv, ; Li Lin, ,
| | - Li Lin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Jiagao Lv, ; Li Lin, ,
| |
Collapse
|
25
|
Brown JS. Treatment of cancer with antipsychotic medications: Pushing the boundaries of schizophrenia and cancer. Neurosci Biobehav Rev 2022; 141:104809. [PMID: 35970416 DOI: 10.1016/j.neubiorev.2022.104809] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/30/2022] [Accepted: 07/31/2022] [Indexed: 10/15/2022]
Abstract
Over a century ago, the phenothiazine dye, methylene blue, was discovered to have both antipsychotic and anti-cancer effects. In the 20th-century, the first phenothiazine antipsychotic, chlorpromazine, was found to inhibit cancer. During the years of elucidating the pharmacology of the phenothiazines, reserpine, an antipsychotic with a long historical background, was likewise discovered to have anti-cancer properties. Research on the effects of antipsychotics on cancer continued slowly until the 21st century when efforts to repurpose antipsychotics for cancer treatment accelerated. This review examines the history of these developments, and identifies which antipsychotics might treat cancer, and which cancers might be treated by antipsychotics. The review also describes the molecular mechanisms through which antipsychotics may inhibit cancer. Although the overlap of molecular pathways between schizophrenia and cancer have been known or suspected for many years, no comprehensive review of the subject has appeared in the psychiatric literature to assess the significance of these similarities. This review fills that gap and discusses what, if any, significance the similarities have regarding the etiology of schizophrenia.
Collapse
|
26
|
Grant CE, Flis A, Ryan BM. Understanding the Role of Dopamine in Cancer: Past, Present, and Future. Carcinogenesis 2022; 43:517-527. [PMID: 35616105 DOI: 10.1093/carcin/bgac045] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 05/12/2022] [Accepted: 05/23/2022] [Indexed: 11/14/2022] Open
Abstract
Dopamine (DA, 3-hydroxytyramine) is member of the catecholamine family and is classically characterized according to its role in the central nervous system as a neurotransmitter. In recent decades, many novel and intriguing discoveries have been made about the peripheral expression of DA receptors (DRs) and the role of DA signaling in both normal and pathological processes. Drawing from decades of evidence suggesting a link between DA and cancer, the DA pathway (DAP) has recently emerged as a potential target in antitumor therapies. Due to the onerous, expensive, and frequently unsuccessful nature of drug development, the repurposing of dopaminergic drugs for cancer therapy has the potential to greatly benefit patients and drug developers alike. However, the lack of clear mechanistic data supporting the direct involvement of DRs and their downstream signaling components in cancer represents an ongoing challenge that has limited the translation of these drugs to the clinic. Despite this, the breadth of evidence linking DA to cancer and non-tumor cells in the tumor microenvironment (TME) justifies further inquiry into the potential applications of this treatment modality in cancer. Herein, we review the literature characterizing the interplay between the DA signaling axis and cancer, highlighting key findings, and then propose rational lines of investigation to follow.
Collapse
Affiliation(s)
- Christopher E Grant
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Amy Flis
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Bríd M Ryan
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
27
|
Chithra NK, Suhas S, Sreeraj VS, Venkatasubramanian G. Clinical utility of trifluoperazine in the treatment of obsessive-compulsive disorder: A case report. Asian J Psychiatr 2022; 71:103051. [PMID: 35245723 DOI: 10.1016/j.ajp.2022.103051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 02/20/2022] [Indexed: 11/02/2022]
Affiliation(s)
- Nellai K Chithra
- Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), India.
| | - Satish Suhas
- Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), India
| | - Vanteemar S Sreeraj
- Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), India
| | | |
Collapse
|
28
|
Jeong JY, Park H, Yoo H, Kim EJ, Jeon B, Lee JD, Kang D, Lee CJ, Paek SH, Roh EJ, Yi GS, Kang SS. Trifluoperazine and Its Analog Suppressed the Tumorigenicity of Non-Small Cell Lung Cancer Cell; Applicability of Antipsychotic Drugs to Lung Cancer Treatment. Biomedicines 2022; 10:biomedicines10051046. [PMID: 35625784 PMCID: PMC9138877 DOI: 10.3390/biomedicines10051046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/21/2022] [Accepted: 04/29/2022] [Indexed: 02/01/2023] Open
Abstract
Despite significant advances in diagnostic and therapeutic technologies, lung cancer remains the leading cause of cancer-related mortality worldwide. Non-small cell lung cancer (NSCLC) accounts for approximately 85% of lung cancer cases. Recently, some antipsychotics have been shown to possess anticancer activity. However, the effects of antipsychotics on NSCLC need to be further explored. We examined the effects of trifluoperazine (TFP), a commonly used antipsychotic drug, and its synthetic analogs on A549 human lung cancer cells. In addition, cell proliferation analysis, colony formation assay, flow cytometry, western blot analysis, and in vivo xenograft experiments were performed. Key genes and mechanisms possibly affected by TFP are significantly related to better survival outcomes in lung cancer patients. Treatment with TFP and a selected TFP analog 3dc significantly inhibited the proliferation, anchorage-dependent/independent colony formation, and migration of A549 cells. Treatment with 3dc affected the expression of genes related to the apoptosis and survival of A549 cells. Treatment with 3dc promoted apoptosis and DNA fragmentation. In all experiments, including in vivo studies of metastatic lung cancer development, 3dc had more substantial anticancer effects than TFP. According to our analysis of publicly available clinical data and in vitro and in vivo experiments, we suggest that some kinds of antipsychotics prevent the progression of NSCLC. Furthermore, this study indicates a synthetic TFP analog that could be a potential therapeutic for lung cancer.
Collapse
Affiliation(s)
- Joo Yeon Jeong
- Department of Anatomy & Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea;
| | - Haangik Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea;
| | - Hong Yoo
- Division of Pulmonology and Allergy, Department of Internal Medicine, Gyeongsang National University Hospital, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (H.Y.); (J.D.L.)
| | - Eun-Jin Kim
- Department of Physiology & Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (E.-J.K.); (D.K.)
| | - Borami Jeon
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Seoul 02792, Korea; (B.J.); (E.J.R.)
| | - Jong Deog Lee
- Division of Pulmonology and Allergy, Department of Internal Medicine, Gyeongsang National University Hospital, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (H.Y.); (J.D.L.)
| | - Dawon Kang
- Department of Physiology & Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (E.-J.K.); (D.K.)
| | - Changjoon Justin Lee
- Center for Glia-Neuron Interaction and Neuroscience, Korea Institute of Science and Technology, Seoul 02792, Korea;
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34141, Korea
| | - Sun Ha Paek
- Department of Neurosurgery, College of Medicine, Seoul National University, Seoul 03080, Korea;
| | - Eun Joo Roh
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Seoul 02792, Korea; (B.J.); (E.J.R.)
| | - Gwan-Su Yi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea;
- Correspondence: (G.-S.Y.); (S.S.K.); Tel.: +82-42-350-4318 (G.-S.Y.); +82-55-772-8033 (S.S.K.)
| | - Sang Soo Kang
- Department of Anatomy & Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea;
- Correspondence: (G.-S.Y.); (S.S.K.); Tel.: +82-42-350-4318 (G.-S.Y.); +82-55-772-8033 (S.S.K.)
| |
Collapse
|
29
|
Lee JS, Kang MJ, Lee JH, Lim DW. Injectable Hydrogels of Stimuli-Responsive Elastin and Calmodulin-Based Triblock Copolypeptides for Controlled Drug Release. Biomacromolecules 2022; 23:2051-2063. [PMID: 35411765 DOI: 10.1021/acs.biomac.2c00053] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A variety of block copolypeptides with stimuli responsiveness have been of growing interest for dynamic self-assembly. Here, multistimuli-responsive triblock copolypeptides composed of thermosensitive elastin-based polypeptides (EBP) and ligand-responsive calmodulin (CalM) were genetically engineered, over-expressed, and nonchromatographically purified by inverse transition cycling. Diluted EBP-CalM-EBP (ECE) triblock copolypeptides under physiological conditions self-assembled into vesicles at the nanoscale by temperature-triggered aggregation of the EBP block with lower critical solution temperature behaviors. Furthermore, concentrated ECE triblock copolypeptides under identical conditions exhibited thermally induced gelation, resulting in physically crosslinked hydrogels. They showed controlled rheological and mechanical properties depending on the conformational change of the CalM middle block induced by binding either Ca2+ or Ca2+ and trifluoperazines (TFPs) as ligands. In addition, both Ca2+-free and Ca2+-bound ECE triblock copolypeptide hydrogels exhibited biocompatibility, while those bound to both Ca2+ and TFPs showed severe cytotoxicity because of controlled TFP release of the CalM blocks. The ECE triblock hydrogels with stimuli responsiveness would be useful as injectable drug delivery depots for biomedical applications.
Collapse
Affiliation(s)
- Jae Sang Lee
- Department of Bionano Engineering and Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Min Jeong Kang
- Department of Bionano Engineering and Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Jae Hee Lee
- Department of Bionano Engineering and Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Dong Woo Lim
- Department of Bionano Engineering and Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| |
Collapse
|
30
|
Singhal S, Maheshwari P, Krishnamurthy PT, Patil VM. Drug Repurposing Strategies for Non-Cancer to Cancer Therapeutics. Anticancer Agents Med Chem 2022; 22:2726-2756. [PMID: 35301945 DOI: 10.2174/1871520622666220317140557] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/15/2021] [Accepted: 11/27/2021] [Indexed: 11/22/2022]
Abstract
Global efforts invested for the prevention and treatment of cancer need to be repositioned to develop safe, effective, and economic anticancer therapeutics by adopting rational approaches of drug discovery. Drug repurposing is one of the established approaches to reposition old, clinically approved off patent noncancer drugs with known targets into newer indications. The literature review suggests key role of drug repurposing in the development of drugs intended for cancer as well as noncancer therapeutics. A wide category of noncancer drugs namely, drugs acting on CNS, anthelmintics, cardiovascular drugs, antimalarial drugs, anti-inflammatory drugs have come out with interesting outcomes during preclinical and clinical phases. In the present article a comprehensive overview of the current scenario of drug repurposing for the treatment of cancer has been focused. The details of some successful studies along with examples have been included followed by associated challenges.
Collapse
Affiliation(s)
- Shipra Singhal
- Department of Pharmaceutical Chemistry KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, India
| | - Priyal Maheshwari
- Department of Pharmaceutical Chemistry KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, India
| | | | - Vaishali M Patil
- Department of Pharmaceutical Chemistry KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, India
| |
Collapse
|
31
|
Chen L, Jiang X, Zhang Q, Li Q, Zhang X, Zhang M, Yu Q, Gao D. How to overcome tumor resistance to anti-PD-1/PD-L1 therapy by immunotherapy modifying the tumor microenvironment in MSS CRC. Clin Immunol 2022; 237:108962. [DOI: 10.1016/j.clim.2022.108962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 12/17/2021] [Accepted: 02/22/2022] [Indexed: 12/21/2022]
|
32
|
Drug Combinations: A New Strategy to Extend Drug Repurposing and Epithelial-Mesenchymal Transition in Breast and Colon Cancer Cells. Biomolecules 2022; 12:biom12020190. [PMID: 35204691 PMCID: PMC8961626 DOI: 10.3390/biom12020190] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 12/21/2022] Open
Abstract
Despite the progressive research and recent advances in drug therapy to treat solid tumours, the number of cases and deaths in patients with cancer is still a major health problem. Drug repurposing coupled to drug combination strategies has been gaining interest among the scientific community. Recently, our group proposed novel drug combinations for breast and colon cancer using repurposed drugs from different classes (antimalarial and central nervous system (CNS)) and chemotherapeutic agents such as 5-fluorouracil (5-FU), paclitaxel (PTX), and found promising results. Here, we proposed a novel drug combination using different CNS drugs and doxorubicin (DOX), an antineoplastic used in breast cancer therapy, and studied their anticancer potential in MCF-7 breast cancer cells. Cells were treated with each drug alone and combined with increasing concentrations of DOX and cell viability was evaluated by MTT and SRB assays. Studies were also complemented with morphological evaluation. Assessment of drug interaction was performed using the CompuSyn and SynergyFinder software. We also compiled our previously studied drug pairs and selected the most promising ones for evaluation of the expression of EMT biomarkers (E-cadherin, P-cadherin, vimentin, and β-catenin) by immunohistochemistry (IHC) to assess if these drug combinations affect the expression of these proteins and eventually revert EMT. These results demonstrate that combination of DOX plus fluoxetine, benztropine, and thioridazine at their IC50 can improve the anticancer effect of DOX but to a lesser degree than when combined with PTX (previous results), resulting in most of the drug interactions being antagonist or additive. This suggests that the choice of the antineoplastic drug influences the success of the drug combination. Collectively, these results also allow us to conclude that antimalarial drugs as repurposed drugs have enhanced effects in MCF-7 breast cancer cells, while combination with CNS drugs seems to be more effective in HT-29 colon cancer cells. The IHC results demonstrate that combination treatments increase E-cadherin expression while reducing P-cadherin, vimentin, and β-catenin, suggesting that these treatments could induce EMT reversal. Taken together, these results could provide promising approaches to the design of novel drug combinations to treat breast and colon cancer patients.
Collapse
|
33
|
Persico M, Abbruzzese C, Matteoni S, Matarrese P, Campana AM, Villani V, Pace A, Paggi MG. Tackling the Behavior of Cancer Cells: Molecular Bases for Repurposing Antipsychotic Drugs in the Treatment of Glioblastoma. Cells 2022; 11:263. [PMID: 35053377 PMCID: PMC8773942 DOI: 10.3390/cells11020263] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/01/2023] Open
Abstract
Glioblastoma (GBM) is associated with a very dismal prognosis, and current therapeutic options still retain an overall unsatisfactorily efficacy in clinical practice. Therefore, novel therapeutic approaches and effective medications are highly needed. Since the development of new drugs is an extremely long, complex and expensive process, researchers and clinicians are increasingly considering drug repositioning/repurposing as a valid alternative to the standard research process. Drug repurposing is also under active investigation in GBM therapy, since a wide range of noncancer and cancer therapeutics have been proposed or investigated in clinical trials. Among these, a remarkable role is played by the antipsychotic drugs, thanks to some still partially unexplored, interesting features of these agents. Indeed, antipsychotic drugs have been described to interfere at variable incisiveness with most hallmarks of cancer. In this review, we analyze the effects of antipsychotics in oncology and how these drugs can interfere with the hallmarks of cancer in GBM. Overall, according to available evidence, mostly at the preclinical level, it is possible to speculate that repurposing of antipsychotics in GBM therapy might contribute to providing potentially effective and inexpensive therapies for patients with this disease.
Collapse
Affiliation(s)
- Michele Persico
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.P.); (C.A.); (S.M.)
| | - Claudia Abbruzzese
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.P.); (C.A.); (S.M.)
| | - Silvia Matteoni
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.P.); (C.A.); (S.M.)
| | - Paola Matarrese
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, 00162 Rome, Italy;
| | - Anna Maria Campana
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA;
| | - Veronica Villani
- Neuro-Oncology, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (V.V.); (A.P.)
| | - Andrea Pace
- Neuro-Oncology, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (V.V.); (A.P.)
| | - Marco G. Paggi
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.P.); (C.A.); (S.M.)
| |
Collapse
|
34
|
Jing Z, Yu W, Li A, Chen X, Chen Y, Chen J. Trifluoperazine Synergistically Potentiates Bortezomib-Induced Anti-Cancer Effect in Multiple Myeloma via Inhibiting P38 MAPK/NUPR1. TOHOKU J EXP MED 2022; 257:315-326. [DOI: 10.1620/tjem.2022.j044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Zizi Jing
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University
| | - Wei Yu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University
| | - Anmao Li
- Department of Respiratory, The First Affiliated Hospital of Chongqing Medical University
| | - Xuanxin Chen
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University
| | - Yuying Chen
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University
| | - Jianbin Chen
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University
| |
Collapse
|
35
|
Hayden E, Holliday H, Lehmann R, Khan A, Tsoli M, Rayner BS, Ziegler DS. Therapeutic Targets in Diffuse Midline Gliomas-An Emerging Landscape. Cancers (Basel) 2021; 13:cancers13246251. [PMID: 34944870 PMCID: PMC8699135 DOI: 10.3390/cancers13246251] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Diffuse midline gliomas (DMGs) remain one of the most devastating childhood brain tumour types, for which there is currently no known cure. In this review we provide a summary of the existing knowledge of the molecular mechanisms underlying the pathogenesis of this disease, highlighting current analyses and novel treatment propositions. Together, the accumulation of these data will aid in the understanding and development of more effective therapeutic options for the treatment of DMGs. Abstract Diffuse midline gliomas (DMGs) are invariably fatal pediatric brain tumours that are inherently resistant to conventional therapy. In recent years our understanding of the underlying molecular mechanisms of DMG tumorigenicity has resulted in the identification of novel targets and the development of a range of potential therapies, with multiple agents now being progressed to clinical translation to test their therapeutic efficacy. Here, we provide an overview of the current therapies aimed at epigenetic and mutational drivers, cellular pathway aberrations and tumor microenvironment mechanisms in DMGs in order to aid therapy development and facilitate a holistic approach to patient treatment.
Collapse
Affiliation(s)
- Elisha Hayden
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington 2052, Australia; (E.H.); (H.H.); (R.L.); (A.K.); (M.T.); (B.S.R.)
| | - Holly Holliday
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington 2052, Australia; (E.H.); (H.H.); (R.L.); (A.K.); (M.T.); (B.S.R.)
- School of Women’s and Children’s Health, Faculty of Medicine, University of New South Wales, Kensington 2052, Australia
| | - Rebecca Lehmann
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington 2052, Australia; (E.H.); (H.H.); (R.L.); (A.K.); (M.T.); (B.S.R.)
- School of Women’s and Children’s Health, Faculty of Medicine, University of New South Wales, Kensington 2052, Australia
| | - Aaminah Khan
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington 2052, Australia; (E.H.); (H.H.); (R.L.); (A.K.); (M.T.); (B.S.R.)
| | - Maria Tsoli
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington 2052, Australia; (E.H.); (H.H.); (R.L.); (A.K.); (M.T.); (B.S.R.)
- School of Women’s and Children’s Health, Faculty of Medicine, University of New South Wales, Kensington 2052, Australia
| | - Benjamin S. Rayner
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington 2052, Australia; (E.H.); (H.H.); (R.L.); (A.K.); (M.T.); (B.S.R.)
- School of Women’s and Children’s Health, Faculty of Medicine, University of New South Wales, Kensington 2052, Australia
| | - David S. Ziegler
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington 2052, Australia; (E.H.); (H.H.); (R.L.); (A.K.); (M.T.); (B.S.R.)
- School of Women’s and Children’s Health, Faculty of Medicine, University of New South Wales, Kensington 2052, Australia
- Kids Cancer Centre, Sydney Children’s Hospital, Randwick 2031, Australia
- Correspondence: ; Tel.: +61-2-9382-1730; Fax: +61-2-9382-1789
| |
Collapse
|
36
|
Repurposing Antipsychotics for Cancer Treatment. Biomedicines 2021; 9:biomedicines9121785. [PMID: 34944601 PMCID: PMC8698939 DOI: 10.3390/biomedicines9121785] [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: 11/09/2021] [Revised: 11/21/2021] [Accepted: 11/24/2021] [Indexed: 11/25/2022] Open
Abstract
Cancer is a leading cause of death worldwide, with approximately 19 million new cases each year. Lately, several novel chemotherapeutic drugs have been introduced, efficiently inhibiting tumor growth and proliferation. However, developing a new drug is a time- and money-consuming process, requiring around 1 billion dollars and nearly ten years, with only a minority of the initially effective anti-cancer drugs experimentally finally being efficient in human clinical trials. Drug repurposing for cancer treatment is an optimal alternative as the safety of these drugs has been previously tested, and thus, in case of successful preclinical studies, can be introduced faster and with a lower cost into phase 3 clinical trials. Antipsychotic drugs are associated with anti-cancer properties and, lately, there has been an increasing interest in their role in cancer treatment. In the present review, we discussed in detail the in-vitro and in-vivo properties of the most common typical and atypical antipsychotics, along with their mechanism of action.
Collapse
|
37
|
Taniguchi K, Suzuki T, Okamura T, Kurita A, Nohara G, Ishii S, Kado S, Takagi A, Tsugane M, Shishido Y. Perifosine, a Bioavailable Alkylphospholipid Akt Inhibitor, Exhibits Antitumor Activity in Murine Models of Cancer Brain Metastasis Through Favorable Tumor Exposure. Front Oncol 2021; 11:754365. [PMID: 34804943 PMCID: PMC8600181 DOI: 10.3389/fonc.2021.754365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/19/2021] [Indexed: 12/18/2022] Open
Abstract
Metastatic brain tumors are regarded as the most advanced stage of certain types of cancer; however, chemotherapy has played a limited role in the treatment of brain metastases. Here, we established murine models of brain metastasis using cell lines derived from human brain metastatic tumors, and aimed to explore the antitumor efficacy of perifosine, an orally active allosteric Akt inhibitor. We evaluated the effectiveness of perifosine by using it as a single agent in ectopic and orthotopic models created by injecting the DU 145 and NCI-H1915 cell lines into mice. Initially, the injected cells formed distant multifocal lesions in the brains of NCI-H1915 mice, making surgical resection impractical in clinical settings. We determined that perifosine could distribute into the brain and remain localized in that region for a long period. Perifosine significantly prolonged the survival of DU 145 and NCI-H1915 orthotopic brain tumor mice; additionally, complete tumor regression was observed in the NCI-H1915 model. Perifosine also elicited much stronger antitumor responses against subcutaneous NCI-H1915 growth; a similar trend of sensitivity to perifosine was also observed in the orthotopic models. Moreover, the degree of suppression of NCI-H1915 tumor growth was associated with long-term exposure to a high level of perifosine at the tumor site and the resultant blockage of the PI3K/Akt signaling pathway, a decrease in tumor cell proliferation, and increased apoptosis. The results presented here provide a promising approach for the future treatment of patients with metastatic brain cancers and emphasize the importance of enriching a patient population that has a higher probability of responding to perifosine.
Collapse
Affiliation(s)
| | - Tomo Suzuki
- Yakult Central Institute, Yakult Honsha Co., Ltd., Tokyo, Japan
| | - Tomomi Okamura
- Yakult Central Institute, Yakult Honsha Co., Ltd., Tokyo, Japan
| | - Akinobu Kurita
- Yakult Central Institute, Yakult Honsha Co., Ltd., Tokyo, Japan
| | - Gou Nohara
- Pharmaceutical Research & Development Department, Yakult Honsha Co., Ltd., Tokyo, Japan
| | - Satoru Ishii
- Pharmaceutical Research & Development Department, Yakult Honsha Co., Ltd., Tokyo, Japan
| | - Shoichi Kado
- Yakult Central Institute, Yakult Honsha Co., Ltd., Tokyo, Japan
| | - Akimitsu Takagi
- Yakult Central Institute, Yakult Honsha Co., Ltd., Tokyo, Japan
| | - Momomi Tsugane
- Yakult Central Institute, Yakult Honsha Co., Ltd., Tokyo, Japan
| | | |
Collapse
|
38
|
Duarte D, Cardoso A, Vale N. Synergistic Growth Inhibition of HT-29 Colon and MCF-7 Breast Cancer Cells with Simultaneous and Sequential Combinations of Antineoplastics and CNS Drugs. Int J Mol Sci 2021; 22:ijms22147408. [PMID: 34299028 PMCID: PMC8306770 DOI: 10.3390/ijms22147408] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 02/06/2023] Open
Abstract
Several central nervous system (CNS) drugs exhibit potent anti-cancer activities. This study aimed to design a novel model of combination that combines different CNS agents and antineoplastic drugs (5-fluorouracil (5-FU) and paclitaxel (PTX)) for colorectal and breast cancer therapy, respectively. Cytotoxic effects of 5-FU and PTX alone and in combination with different CNS agents were evaluated on HT-29 colon and MCF-7 breast cancer cells, respectively. Three antimalarials alone and in combination with 5-FU were also evaluated in HT-29 cells. Different schedules and concentrations in a fixed ratio were added to the cultured cells and incubated for 48 h. Cell viability was evaluated using MTT and SRB assays. Synergism was evaluated using the Chou-Talalay, Bliss Independence and HSA methods. Our results demonstrate that fluphenazine, fluoxetine and benztropine have enhanced anticancer activity when used alone as compared to being used in combination, making them ideal candidates for drug repurposing in colorectal cancer (CRC). Regarding MCF-7 cells, sertraline was the most promising candidate alone for drug repurposing, with the lowest IC50 value. For HT-29 cells, the CNS drugs sertraline and thioridazine in simultaneous combination with 5-FU demonstrated the strongest synergism among all combinations. In MCF-7 breast cancer cells, the combination of fluoxetine, fluphenazine and benztropine with PTX resulted in synergism for all concentrations below IC50. We also found that the antimalarial artesunate administration prior to 5-FU produces better results in reducing HT-29 cell viability than the inverse drug schedule or the simultaneous combination. These results demonstrate that CNS drugs activity differs between the two selected cell lines, both alone and in combination, and support that some CNS agents may be promising candidates for drug repurposing in these types of cancers. Additionally, these results demonstrate that 5-FU or a combination of PTX with CNS drugs should be further evaluated. These results also demonstrate that antimalarial drugs may also be used as antitumor agents in colorectal cancer, besides breast cancer.
Collapse
Affiliation(s)
- Diana Duarte
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Armando Cardoso
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Correspondence:
| |
Collapse
|
39
|
Cui C, Zhang Y, Liu G, Zhang S, Zhang J, Wang X. Advances in the study of cancer metastasis and calcium signaling as potential therapeutic targets. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:266-291. [PMID: 36046433 PMCID: PMC9400724 DOI: 10.37349/etat.2021.00046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/21/2021] [Indexed: 11/19/2022] Open
Abstract
Metastasis is still the primary cause of cancer-related mortality. However, the underlying mechanisms of cancer metastasis are not yet fully understood. Currently, the epithelial-mesenchymal transition, metabolic remodeling, cancer cell intercommunication and the tumor microenvironment including diverse stromal cells, are reported to affect the metastatic process of cancer cells. Calcium ions (Ca2+) are ubiquitous second messengers that manipulate cancer metastasis by affecting signaling pathways. Diverse transporter/pump/channel-mediated Ca2+ currents form Ca2+ oscillations that can be decoded by Ca2+-binding proteins, which are promising prognostic biomarkers and therapeutic targets of cancer metastasis. This paper presents a review of the advances in research on the mechanisms underlying cancer metastasis and the roles of Ca2+-related signals in these events.
Collapse
Affiliation(s)
- Chaochu Cui
- Henan Key Laboratory of Medical Tissue Regeneration, College of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Yongxi Zhang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Gang Liu
- Henan Key Laboratory of Medical Tissue Regeneration, College of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Shuhong Zhang
- Henan Key Laboratory of Medical Tissue Regeneration, College of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Jinghang Zhang
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, College of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| |
Collapse
|
40
|
So JS, Kim H, Han KS. Mechanisms of Invasion in Glioblastoma: Extracellular Matrix, Ca 2+ Signaling, and Glutamate. Front Cell Neurosci 2021; 15:663092. [PMID: 34149360 PMCID: PMC8206529 DOI: 10.3389/fncel.2021.663092] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most common and malignant form of primary brain tumor with a median survival time of 14–16 months in GBM patients. Surgical treatment with chemotherapy and radiotherapy may help increase survival by removing GBM from the brain. However, complete surgical resection to eliminate GBM is almost impossible due to its high invasiveness. When GBM cells migrate to the brain, they interact with various cells, including astrocytes, neurons, endothelial cells, and the extracellular matrix (ECM). They can also make their cell body shrink to infiltrate into narrow spaces in the brain; thereby, they can invade regions of the brain and escape from surgery. Brain tumor cells create an appropriate microenvironment for migration and invasion by modifying and degrading the ECM. During those processes, the Ca2+ signaling pathway and other signaling cascades mediated by various ion channels contribute mainly to gene expression, motility, and invasion of GBM cells. Furthermore, GBM cells release glutamate, affecting migration via activation of ionotropic glutamate receptors in an autocrine manner. This review focuses on the cellular mechanisms of glioblastoma invasion and motility related to ECM, Ca2+ signaling, and glutamate. Finally, we discuss possible therapeutic interventions to inhibit invasion by GBM cells.
Collapse
Affiliation(s)
- Jae-Seon So
- Department of Medical Biotechnology, Dongguk University-Gyeongju, Gyeongju, South Korea
| | - Hyeono Kim
- Department of Medical Biotechnology, Dongguk University-Gyeongju, Gyeongju, South Korea
| | - Kyung-Seok Han
- Department of Medical Biotechnology, Dongguk University-Gyeongju, Gyeongju, South Korea
| |
Collapse
|
41
|
Wongdee K, Chanpaisaeng K, Teerapornpuntakit J, Charoenphandhu N. Intestinal Calcium Absorption. Compr Physiol 2021; 11:2047-2073. [PMID: 34058017 DOI: 10.1002/cphy.c200014] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In this article, we focus on mammalian calcium absorption across the intestinal epithelium in normal physiology. Intestinal calcium transport is essential for supplying calcium for metabolism and bone mineralization. Dietary calcium is transported across the mucosal epithelia via saturable transcellular and nonsaturable paracellular pathways, both of which are under the regulation of 1,25-dihydroxyvitamin D3 and several other endocrine and paracrine factors, such as parathyroid hormone, prolactin, 17β-estradiol, calcitonin, and fibroblast growth factor-23. Calcium absorption occurs in several segments of the small and large intestine with varying rates and capacities. Segmental heterogeneity also includes differential expression of calcium transporters/carriers (e.g., transient receptor potential cation channel and calbindin-D9k ) and the presence of favorable factors (e.g., pH, luminal contents, and gut motility). Other proteins and transporters (e.g., plasma membrane vitamin D receptor and voltage-dependent calcium channels), as well as vesicular calcium transport that probably contributes to intestinal calcium absorption, are also discussed. © 2021 American Physiological Society. Compr Physiol 11:1-27, 2021.
Collapse
Affiliation(s)
- Kannikar Wongdee
- Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand.,Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Krittikan Chanpaisaeng
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand.,Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Jarinthorn Teerapornpuntakit
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand.,Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Narattaphol Charoenphandhu
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand.,Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand.,Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand.,The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| |
Collapse
|
42
|
Antoszczak M, Markowska A, Markowska J, Huczyński A. Antidepressants and Antipsychotic Agents as Repurposable Oncological Drug Candidates. Curr Med Chem 2021; 28:2137-2174. [PMID: 32895037 DOI: 10.2174/0929867327666200907141452] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/26/2020] [Accepted: 06/10/2020] [Indexed: 11/22/2022]
Abstract
Drug repurposing, also known as drug repositioning/reprofiling, is a relatively new strategy for the identification of alternative uses of well-known therapeutics that are outside the scope of their original medical indications. Such an approach might entail a number of advantages compared to standard de novo drug development, including less time needed to introduce the drug to the market, and lower costs. The group of compounds that could be considered as promising candidates for repurposing in oncology include the central nervous system drugs, especially selected antidepressant and antipsychotic agents. In this article, we provide an overview of some antidepressants (citalopram, fluoxetine, paroxetine, sertraline) and antipsychotics (chlorpromazine, pimozide, thioridazine, trifluoperazine) that have the potential to be repurposed as novel chemotherapeutics in cancer treatment, as they have been found to exhibit preventive and/or therapeutic action in cancer patients. Nevertheless, although drug repurposing seems to be an attractive strategy to search for oncological drugs, we would like to clearly indicate that it should not replace the search for new lead structures, but only complement de novo drug development.
Collapse
Affiliation(s)
- Michał Antoszczak
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Poznan, Poland
| | - Anna Markowska
- \Department of Perinatology and Women's Diseases, Poznań University of Medical Sciences, Poznan, Poland
| | - Janina Markowska
- Department of Oncology, Poznań University of Medical Sciences, Poznan, Poland
| | - Adam Huczyński
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Poznan, Poland
| |
Collapse
|
43
|
Bustos G, Ahumada-Castro U, Silva-Pavez E, Puebla A, Lovy A, Cesar Cardenas J. The ER-mitochondria Ca 2+ signaling in cancer progression: Fueling the monster. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 363:49-121. [PMID: 34392932 DOI: 10.1016/bs.ircmb.2021.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer is a leading cause of death worldwide. All major tumor suppressors and oncogenes are now recognized to have fundamental connections with metabolic pathways. A hallmark feature of cancer cells is a reprogramming of their metabolism even when nutrients are available. Increasing evidence indicates that most cancer cells rely on mitochondrial metabolism to sustain their energetic and biosynthetic demands. Mitochondria are functionally and physically coupled to the endoplasmic reticulum (ER), the major calcium (Ca2+) storage organelle in mammalian cells, through special domains known as mitochondria-ER contact sites (MERCS). In this domain, the release of Ca2+ from the ER is mainly regulated by inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs), a family of Ca2+ release channels activated by the ligand IP3. IP3R mediated Ca2+ release is transferred to mitochondria through the mitochondrial Ca2+ uniporter (MCU). Once in the mitochondrial matrix, Ca2+ activates several proteins that stimulate mitochondrial performance. The role of IP3R and MCU in cancer, as well as the other proteins that enable the Ca2+ communication between these two organelles is just beginning to be understood. Here, we describe the function of the main players of the ER mitochondrial Ca2+ communication and discuss how this particular signal may contribute to the rise and development of cancer traits.
Collapse
Affiliation(s)
- Galdo Bustos
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Ulises Ahumada-Castro
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Eduardo Silva-Pavez
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Andrea Puebla
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Alenka Lovy
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Department of Neuroscience, Center for Neuroscience Research, Tufts School of Medicine, Boston, MA, United States.
| | - J Cesar Cardenas
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, United States; Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA, United States.
| |
Collapse
|
44
|
Wang B, Zhong JL, Li HZ, Wu B, Sun DF, Jiang N, Shang J, Chen YF, Xu XH, Lu HD. Diagnostic and therapeutic values of PMEPA1 and its correlation with tumor immunity in pan-cancer. Life Sci 2021; 277:119452. [PMID: 33831430 DOI: 10.1016/j.lfs.2021.119452] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 12/30/2022]
Abstract
AIMS The prostate transmembrane protein, androgen induced 1 (PMEPA1) is differentially expressed in pan-cancer. However, PMEPA1 specific role in cancers has not been fully clarified. This study aims to explore the potential role of Pmepa1 in pan-cancer and specific cancer, with a view to deepening the research on the pathological mechanism of cancer. MAIN METHODS The Perl language and R language were used to identify the correlation between PMEPA1 expression level and clinical indicators, prognosis values, tumor microenvironment, immune cells' infiltration, immune checkpoint genes, TMB and MSI. The Therapeutic Target Database was used for identifying potential therapeutic drugs that target the pathways that are significantly affected by PMEPA1 expression. KEY FINDINGS PMEPA1 differential expression significantly correlated with patients' age, race, tumors' stage and status. PMEPA1 high expression was closely correlated with poor prognosis in many cancer types, excluding prostate adenocarcinoma. PMEPA1 expression was closely related to tumor cells and the immune microenvironment in stromal and immune cells' level, immune cells' infiltration, immune checkpoint genes, tumor mutational burden and microsatellite instability. We also found that the activity of the olfactory transduction pathway was closely related to PMEPA1 expression. In pan-cancer, Trifluoperazine and Halofantrine have the potential to reduce PMEPA1 expression. SIGNIFICANCE This study integrated existing data to explore PMEPA1 potential function in cancers, provided insights for the future cancer-related studies.
Collapse
Affiliation(s)
- Bin Wang
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China
| | - Jun-Long Zhong
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China
| | - Hui-Zi Li
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China
| | - Biao Wu
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China
| | - Di-Fang Sun
- Department of Ophthalmology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
| | - Ning Jiang
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China
| | - Jie Shang
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China
| | - Yu-Feng Chen
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China
| | - Xiang-He Xu
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China.
| | - Hua-Ding Lu
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China.
| |
Collapse
|
45
|
Preclinical Investigation of Trifluoperazine as a Novel Therapeutic Agent for the Treatment of Pulmonary Arterial Hypertension. Int J Mol Sci 2021; 22:ijms22062919. [PMID: 33805714 PMCID: PMC7998101 DOI: 10.3390/ijms22062919] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/17/2022] Open
Abstract
Trifluoperazine (TFP), an antipsychotic drug approved by the Food and Drug Administration, has been show to exhibit anti-cancer effects. Pulmonary arterial hypertension (PAH) is a devastating disease characterized by a progressive obliteration of small pulmonary arteries (PAs) due to exaggerated proliferation and resistance to apoptosis of PA smooth muscle cells (PASMCs). However, the therapeutic potential of TFP for correcting the cancer-like phenotype of PAH-PASMCs and improving PAH in animal models remains unknown. PASMCs isolated from PAH patients were exposed to different concentrations of TFP before assessments of cell proliferation and apoptosis. The in vivo therapeutic potential of TFP was tested in two preclinical models with established PAH, namely the monocrotaline and sugen/hypoxia-induced rat models. Assessments of hemodynamics by right heart catheterization and histopathology were conducted. TFP showed strong anti-survival and anti-proliferative effects on cultured PAH-PASMCs. Exposure to TFP was associated with downregulation of AKT activity and nuclear translocation of forkhead box protein O3 (FOXO3). In both preclinical models, TFP significantly lowered the right ventricular systolic pressure and total pulmonary resistance and improved cardiac function. Consistently, TFP reduced the medial wall thickness of distal PAs. Overall, our data indicate that TFP could have beneficial effects in PAH and support the view that seeking new uses for old drugs may represent a fruitful approach.
Collapse
|
46
|
Ahumada-Castro U, Bustos G, Silva-Pavez E, Puebla-Huerta A, Lovy A, Cárdenas C. In the Right Place at the Right Time: Regulation of Cell Metabolism by IP3R-Mediated Inter-Organelle Ca 2+ Fluxes. Front Cell Dev Biol 2021; 9:629522. [PMID: 33738285 PMCID: PMC7960657 DOI: 10.3389/fcell.2021.629522] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 01/19/2021] [Indexed: 12/18/2022] Open
Abstract
In the last few years, metabolism has been shown to be controlled by cross-organelle communication. The relationship between the endoplasmic reticulum and mitochondria/lysosomes is the most studied; here, inositol 1,4,5-triphosphate (IP3) receptor (IP3R)-mediated calcium (Ca2+) release plays a central role. Recent evidence suggests that IP3R isoforms participate in synthesis and degradation pathways. This minireview will summarize the current findings in this area, emphasizing the critical role of Ca2+ communication on organelle function as well as catabolism and anabolism, particularly in cancer.
Collapse
Affiliation(s)
- Ulises Ahumada-Castro
- Geroscience Center for Brain Health and Metabolism, Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Galdo Bustos
- Geroscience Center for Brain Health and Metabolism, Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Eduardo Silva-Pavez
- Geroscience Center for Brain Health and Metabolism, Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Andrea Puebla-Huerta
- Geroscience Center for Brain Health and Metabolism, Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Alenka Lovy
- Geroscience Center for Brain Health and Metabolism, Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.,Department of Neuroscience, Center for Neuroscience Research, Tufts University School of Medicine, Boston, MA, United States
| | - César Cárdenas
- Geroscience Center for Brain Health and Metabolism, Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.,Buck Institute for Research on Aging, Novato, CA, United States.,Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|
47
|
Leal NS, Martins LM. Mind the Gap: Mitochondria and the Endoplasmic Reticulum in Neurodegenerative Diseases. Biomedicines 2021; 9:biomedicines9020227. [PMID: 33672391 PMCID: PMC7926795 DOI: 10.3390/biomedicines9020227] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/16/2022] Open
Abstract
The way organelles are viewed by cell biologists is quickly changing. For many years, these cellular entities were thought to be unique and singular structures that performed specific roles. However, in recent decades, researchers have discovered that organelles are dynamic and form physical contacts. In addition, organelle interactions modulate several vital biological functions, and the dysregulation of these contacts is involved in cell dysfunction and different pathologies, including neurodegenerative diseases. Mitochondria–ER contact sites (MERCS) are among the most extensively studied and understood juxtapositioned interorganelle structures. In this review, we summarise the major biological and ultrastructural dysfunctions of MERCS in neurodegeneration, with a particular focus on Alzheimer’s disease as well as Parkinson’s disease, amyotrophic lateral sclerosis and frontotemporal dementia. We also propose an updated version of the MERCS hypothesis in Alzheimer’s disease based on new findings. Finally, we discuss the possibility of MERCS being used as possible drug targets to halt cell death and neurodegeneration.
Collapse
|
48
|
Tilak M, Holborn J, New LA, Lalonde J, Jones N. Receptor Tyrosine Kinase Signaling and Targeting in Glioblastoma Multiforme. Int J Mol Sci 2021; 22:1831. [PMID: 33673213 PMCID: PMC7918566 DOI: 10.3390/ijms22041831] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma multiforme (GBM) is amongst the deadliest of human cancers, with a median survival rate of just over one year following diagnosis. Characterized by rapid proliferation and diffuse infiltration into the brain, GBM is notoriously difficult to treat, with tumor cells showing limited response to existing therapies and eventually developing resistance to these interventions. As such, there is intense interest in better understanding the molecular alterations in GBM to guide the development of more efficient targeted therapies. GBM tumors can be classified into several molecular subtypes which have distinct genetic signatures, and they show aberrant activation of numerous signal transduction pathways, particularly those connected to receptor tyrosine kinases (RTKs) which control glioma cell growth, survival, migration, invasion, and angiogenesis. There are also non-canonical modes of RTK signaling found in GBM, which involve G-protein-coupled receptors and calcium channels. This review uses The Cancer Genome Atlas (TCGA) GBM dataset in combination with a data-mining approach to summarize disease characteristics, with a focus on select molecular pathways that drive GBM pathogenesis. We also present a unique genomic survey of RTKs that are frequently altered in GBM subtypes, as well as catalog the GBM disease association scores for all RTKs. Lastly, we discuss current RTK targeted therapies and highlight emerging directions in GBM research.
Collapse
Affiliation(s)
| | | | | | | | - Nina Jones
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada; (M.T.); (J.H.); (L.A.N.); (J.L.)
| |
Collapse
|
49
|
Sylvain NJ, Salman MM, Pushie MJ, Hou H, Meher V, Herlo R, Peeling L, Kelly ME. The effects of trifluoperazine on brain edema, aquaporin-4 expression and metabolic markers during the acute phase of stroke using photothrombotic mouse model. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183573. [PMID: 33561476 DOI: 10.1016/j.bbamem.2021.183573] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/23/2021] [Accepted: 01/29/2021] [Indexed: 12/22/2022]
Abstract
Stroke is the second leading cause of death and the third leading cause of disability globally. Edema is a hallmark of stroke resulting from dysregulation of water homeostasis in the central nervous system (CNS) and plays the major role in stroke-associated morbidity and mortality. The overlap between cellular and vasogenic edema makes treating this condition complicated, and to date, there is no pathogenically oriented drug treatment for edema. Water balance in the brain is tightly regulated, primarily by aquaporin 4 (AQP4) channels, which are mainly expressed in perivascular astrocytic end-feet. Targeting AQP4 could be a useful therapeutic approach for treating brain edema; however, there is no approved drug for stroke treatment that can directly block AQP4. In this study, we demonstrate that the FDA-approved drug trifluoperazine (TFP) effectively reduces cerebral edema during the early acute phase in post-stroke mice using a photothrombotic stroke model. This effect was combined with an inhibition of AQP4 expression at gene and protein levels. Importantly, TFP does not appear to induce any deleterious changes on brain electrolytes or metabolic markers, including total protein or lipid levels. Our results support a possible role for TFP in providing a beneficial extra-osmotic effect on brain energy metabolism, as indicated by the increase of glycogen levels. We propose that targeting AQP4-mediated brain edema using TFP is a viable therapeutic strategy during the early and acute phase of stroke that can be further investigated during later stages to help in developing novel CNS edema therapies.
Collapse
Affiliation(s)
- Nicole J Sylvain
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Canada
| | - Mootaz M Salman
- Medical Sciences Division, Department of Physiology, Anatomy and Genetics, Oxford University, South Parks Road, Oxford OX1 3QX, UK.
| | - M Jake Pushie
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Canada
| | - Huishu Hou
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Canada
| | - Vedashree Meher
- Department of Anatomy and Cell Biology, College of Medicine University of Saskatchewan, Canada
| | - Rasmus Herlo
- Neurotechnology Center, Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Lissa Peeling
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Canada
| | - Michael E Kelly
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Canada
| |
Collapse
|
50
|
Hsu WC, Le HN, Lin YJ, Chen MC, Wang TF, Li CC, Kuo WW, Mahalakshmi B, Singh CH, Chen MC, Huang CY. Calmodulin/CaMKII-γ mediates prosurvival capability in apicidin-persistent hepatocellular carcinoma cells via ERK1/2/CREB/c-fos signaling pathway. J Cell Biochem 2021; 122:612-625. [PMID: 33459431 DOI: 10.1002/jcb.29892] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/27/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023]
Abstract
Calmodulin (CaM), a Ca2+ binding protein, plays a critical role in cancer initiation and progression through binding and activating numerous target proteins, including Ca2+ /calmodulin-dependent protein kinase (CaMK) family proteins. However, the mechanisms underlying the effects of CaM/CaMKs on the survival capability of liver cancer cells is unclear, and this study investigates this mechanism in apicidin-persistent HA22T cells. CaM level was upregulated, especially in the cytosol, in apicidin-persistent HA22T cells than in parental HA22T cells and was positively associated with cell proliferation and migration capacity of apicidin-persistent HA22T cells. Further, the expression of CaM-activated CaMKs-dependent signaling cascades, including CaMKK2, CaMKIV, CaMKII-γ, and p-CaMKII was observed in apicidin-persistent HA22T cells, which were transiently activated by mitogen-activated protein kinase oncogenic signaling, such as CREB, ERK1/2, and c-fos. Furthermore, a specific CaM inhibitor trifluoperazine reduced the levels of p-CREB, p-ERK1/2, and c-fos in apicidin-persistent HA22T cells than in parental HA22T cells. Additionally, inhibition of CaM also suppressed CaM-induced Bcl-XL (an antiapoptotic protein) expression in apicidin-persistent HA22T cells. Our finding emphasizes an essential role of CaM/CaMKs in augmentation of the survival capability of apicidin-persistent liver cancer cells and suggests that CaM inhibition significantly attenuates CaM-induced tumor growth and abrogates antiapoptotic function and also offers a promising therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Wei-Chung Hsu
- Department of Radiation Oncology, Chung-Kang Branch, Cheng-Ching General Hospital, Taichung, Taiwan.,Department of Occupational Therapy, Asia University, Taichung, Taiwan
| | - Hang-Nga Le
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Yu-Jung Lin
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ming-Cheng Chen
- Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Tso-Fu Wang
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chi-Cheng Li
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Center of Stem Cell & Precision Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - B Mahalakshmi
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
| | - Chaouhan Hitesh Singh
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Mei-Chih Chen
- Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan.,Department of Nursing, Asia University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Department of Biological Science and Technology, Asia University, Taichung, Taiwan.,Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|