1
|
Mugisha S, Baba SA, Labhsetwar S, Dave D, Zakeri A, Klemke R, Desgrosellier JS. S100A8/A9 innate immune signaling as a distinct mechanism driving progression of smoking-related breast cancers. Oncogene 2025:10.1038/s41388-025-03276-5. [PMID: 39856330 DOI: 10.1038/s41388-025-03276-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Smoking plays an underappreciated role in breast cancer progression, increasing recurrence and mortality in patients. Here, we show that S100A8/A9 innate immune signaling is a molecular mechanism that identifies smoking-related breast cancers and underlies their enhanced malignancy. In contrast to acute exposure, chronic nicotine increased tumorigenicity and reprogrammed breast cancer cells to express innate immune response genes. This required the α7 nicotinic acetylcholine receptor, which elicited dynamic changes in cell differentiation, proliferation, and expression of secreted cytokines, such as S100A8 and S100A9, as assessed by unbiased scRNA-seq. Indeed, pharmacologic or genetic inhibition of S100A8/A9-RAGE receptor signaling blocked nicotine's tumor-promoting effects. We also discovered Syntaphilin (SNPH) as an S100A8/A9-dependent gene enriched specifically in estrogen receptor-negative (ER-) cancers from former smokers, linking this response to patient disease. Together, our findings describe a new α7 nAChR-S100A8/A9-Syntaphilin immune signaling module that drives nicotine-induced tumor progression and distinguishes smoking-related patient disease as a distinct subset of aggressive breast cancers.
Collapse
Affiliation(s)
- Samson Mugisha
- Department of Pathology, University of California, San Diego, La Jolla, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, USA
| | - Shahnawaz A Baba
- Department of Pathology, University of California, San Diego, La Jolla, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, USA
| | - Shreyas Labhsetwar
- Department of Pathology, University of California, San Diego, La Jolla, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, USA
| | - Devam Dave
- Department of Pathology, University of California, San Diego, La Jolla, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, USA
| | - Aran Zakeri
- Department of Pathology, University of California, San Diego, La Jolla, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, USA
| | - Richard Klemke
- Department of Pathology, University of California, San Diego, La Jolla, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, USA
| | - Jay S Desgrosellier
- Department of Pathology, University of California, San Diego, La Jolla, USA.
- Moores Cancer Center, University of California, San Diego, La Jolla, USA.
| |
Collapse
|
2
|
Wang Q, Zhao X, Wang S, Lu S. Sarcopenia and immune-mediated inflammatory diseases: Evaluating causality and exploring therapeutic targets for sarcopenia through Mendelian randomization. Int Immunopharmacol 2025; 144:113687. [PMID: 39591827 DOI: 10.1016/j.intimp.2024.113687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/03/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND An increasing body of evidence has revealed the association between immune-mediated inflammatory diseases (IMIDs) and sarcopenia. However, a genetically direct causality between IMIDs and sarcopenia remains elusive. METHODS To investigate the relationship between IMIDs and sarcopenia-related traits and identify potential therapeutic targets, a Mendelian randomization (MR) was performed. We collected publicly available genome-wide association studies (GWAS) data for seven common IMIDs, including systemic lupus erythematosus (SLE), inflammatory bowel disease (IBD), Crohn's disease (CD), ulcerative colitis (UC), psoriasis (PSO), ankylosing spondylitis (AS), and rheumatoid arthritis (RA). Additionally, summary-level GWAS data for sarcopenia-related traits, including appendicular lean mass (ALM), left-hand grip strength, and right-hand grip strength were collected. To search for therapeutic targets, we used two types of genetic instruments to proxy the exposure of druggable genes, including genetic variants within or nearby drug targets and expression quantitative trait loci (eQTLs) of drug targets. Two-sample MR and summary-data-based MR (SMR) were used to calculate effect estimates, and sensitivity analyses were implemented for robustness. Drug tractability, gene enrichment analysis, and protein-protein interaction (PPI) analysis were used to validate the biological and clinical significance of the selected drug targets. RESULTS The two-sample MR analysis indicated the existence of casual associations between IMIDs and sarcopenia-related traits in the overall and sex-stratified populations. In particular, PSO had causal effects on decreased ALM, which showed significance in all six MR analysis tests with directional consistency in the overall population. Grounded in this robust association, HLA-DRB5, HLA-DRB1, and AGER were identified as potential therapeutic targets for ALM decline by drug target MR and further confirmed by SMR analysis. These genes were associated with therapeutic agents currently undergoing evaluations in clinical trials. Gene enrichment and PPI analysis indicated a strong association of these genes with immune functions. CONCLUSIONS This MR study contributes novel genetic evidence supporting the causal link between IMIDs and sarcopenia, with a particular emphasis on the association between PSO and decreased ALM. Additionally, AGER, HLA-DRB1, and HLA-DRB5 emerge as potential therapeutic targets for ALM decline.
Collapse
Affiliation(s)
- Qijun Wang
- Department of Orthopedics & Elderly Spinal Surgery, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Xuan Zhao
- Department of Orthopedics & Elderly Spinal Surgery, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Shuaikang Wang
- Department of Orthopedics & Elderly Spinal Surgery, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Shibao Lu
- Department of Orthopedics & Elderly Spinal Surgery, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China.
| |
Collapse
|
3
|
Coser M, Neamtu BM, Pop B, Cipaian CR, Crisan M. RAGE and its ligands in breast cancer progression and metastasis. Oncol Rev 2025; 18:1507942. [PMID: 39830522 PMCID: PMC11739297 DOI: 10.3389/or.2024.1507942] [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: 10/08/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Breast cancer is the most common form of cancer diagnosed worldwide and the leading cause of death in women globally, according to Globocan 2020. Hence, investigating novel pathways implicated in cancer progression and metastasis could lead to the development of targeted therapies and new treatment strategies in breast cancer. Recent studies reported an interplay between the receptor for advanced glycation end products (RAGE) and its ligands, S100 protein group, advanced glycation end products (AGEs) and high-mobility group box 1 protein (HMGB1) and breast cancer growth and metastasis. Materials and methods We used articles available in the NCBI website database PubMed to write this scoping review. The search words used were 'RAGE receptor' AND/OR 'breast cancer, RAGE ligands, glycation end products'. A total of 90 articles were included. We conducted a meta-analysis to assess the relationship between the RAGE rs1800624 polymorphism and breast cancer risk using fixed-effect or random-effect models to calculate odds ratios (ORs) and their corresponding 95% confidence intervals (95% CIs). Results RAGE upon activation by its ligands enhances downstream signaling pathways, contributing to breast cancer cells migration, growth, angiogenesis, metastasis, and drug resistance. In addition, studies have shown that RAGE and its ligands influence the way breast cancer cells interact with immune cells present in the tumor microenvironment (macrophages, fibroblasts), thus regulating it to promote tumor growth and metastasis. Conclusion Breast cancers with a high expression of RAGE are associated with poor prognosis. Targeting RAGE and its ligands impairs cell invasion and metastasis, showing promising potential for further research as potential prognostic biomarkers or targeted onco-therapeutics.
Collapse
Affiliation(s)
- Madalina Coser
- Department of Histology, Doctoral School “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
| | - Bogdan Mihai Neamtu
- Clinical Medical Department, Center for Research in Mathematics and Applications, Faculty of Medicine, “Lucian Blaga” University Sibiu, Sibiu, Romania
- Department of Clinical Research, Pediatric Clinical Hospital Sibiu, Sibiu, Romania
| | - Bogdan Pop
- Department of Pathology, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
- Department of Pathology, “Prof. Dr. ion Chiricuta” Institute of Oncology Cluj-Napoca, Cluj-Napoca, Romania
| | - Calin Remus Cipaian
- Second Medical Clinic, Sibiu County Clinical Emergency Hospital, Sibiu, Romania
- Clinical Medical Department, Faculty of Medicine, “Lucian Blaga” University Sibiu, Sibiu, Romania
| | - Maria Crisan
- Department of Histology, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
- Clinic of Dermatology, Emergency Clinical County Hospital, Cluj-Napoca, Romania
| |
Collapse
|
4
|
Alka K, Oyeniyi JF, Mohammad G, Zhao Y, Marcus S, Chinnaiyan P. The RAGE Inhibitor TTP488 (Azeliragon) Demonstrates Anti-Tumor Activity and Enhances the Efficacy of Radiation Therapy in Pancreatic Cancer Cell Lines. Cancers (Basel) 2024; 17:17. [PMID: 39796649 PMCID: PMC11718873 DOI: 10.3390/cancers17010017] [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: 11/28/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
Pancreatic cancer is the third leading cause of cancer-related mortality in the United States, with rising incidence and mortality. The receptor for advanced glycation end products (RAGE) and its ligands significantly contribute to pancreatic cancer progression by enhancing cell proliferation, fostering treatment resistance, and promoting a pro-tumor microenvironment via activation of the nuclear factor-kappa B (NF-κB) signaling pathways. This study validated pathway activation in human pancreatic cancer and evaluated the therapeutic efficacy of TTP488 (Azeliragon), a small-molecule RAGE inhibitor, alone and in combination with radiation therapy (RT) in preclinical models of pancreatic cancer. Human (Panc1) and murine (Pan02) pancreatic cancer cell lines exhibited elevated levels of RAGE and its ligands compared to normal pancreatic tissue. In vitro, Azeliragon inhibited RAGE-mediated NF-κB activation and ligand-mediated cell proliferation in pancreatic cancer cell lines. Target engagement of Azeliragon was confirmed in vivo, as determined by decreased NF-κB activation. Azeliragon demonstrated significant growth delay in mouse models of pancreatic cancer and additive effects when combined with RT. Additionally, Azeliragon modulated the immune suppressive tumor microenvironment in pancreatic cancer by reducing immunosuppressive cells, including M2 macrophages, regulatory T cells, and myeloid-derived suppressor cells, while enhancing CD8+ T cell infiltration. These findings suggest that Azeliragon, by inhibiting RAGE-mediated signaling and modulating immune response, may serve as an effective anti-cancer agent in pancreatic cancer.
Collapse
Affiliation(s)
- Kumari Alka
- Department of Radiation Oncology, Corewell Health William Beaumont University Hospital, Royal Oak, MI 48076, USA; (K.A.); (J.F.O.); (G.M.); (Y.Z.)
| | - Jacob F. Oyeniyi
- Department of Radiation Oncology, Corewell Health William Beaumont University Hospital, Royal Oak, MI 48076, USA; (K.A.); (J.F.O.); (G.M.); (Y.Z.)
| | - Ghulam Mohammad
- Department of Radiation Oncology, Corewell Health William Beaumont University Hospital, Royal Oak, MI 48076, USA; (K.A.); (J.F.O.); (G.M.); (Y.Z.)
| | - Yi Zhao
- Department of Radiation Oncology, Corewell Health William Beaumont University Hospital, Royal Oak, MI 48076, USA; (K.A.); (J.F.O.); (G.M.); (Y.Z.)
| | | | - Prakash Chinnaiyan
- Department of Radiation Oncology, Corewell Health William Beaumont University Hospital, Royal Oak, MI 48076, USA; (K.A.); (J.F.O.); (G.M.); (Y.Z.)
- Radiation Oncology, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| |
Collapse
|
5
|
Sigalov AB. TREM-1 and TREM-2 as therapeutic targets: clinical challenges and perspectives. Front Immunol 2024; 15:1498993. [PMID: 39737196 PMCID: PMC11682994 DOI: 10.3389/fimmu.2024.1498993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/30/2024] [Indexed: 01/01/2025] Open
Abstract
TREM-1 and TREM-2 as Therapeutic Targets: Clinical Challenges and Perspectives.
Collapse
|
6
|
Hayat C, Yaseen M, Ahmad S, Khalid K, Alamri MA, Khalid A, Shah SQ, Ejiohuo O, Wadood A, Maigoro AY, Kwon HW. Elucidating the interactions of advanced glycation end products with RAGE, employing molecular docking and MD simulation approaches: Implications of potent therapeutic for diabetes and its related complications. J Mol Liq 2024; 416:126467. [DOI: 10.1016/j.molliq.2024.126467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
7
|
Sarkar S. Pathological role of RAGE underlying progression of various diseases: its potential as biomarker and therapeutic target. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03595-6. [PMID: 39589529 DOI: 10.1007/s00210-024-03595-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/31/2024] [Indexed: 11/27/2024]
Abstract
The receptor for advanced glycation end products (RAGE) is a multi-ligand receptor with several structural types, performing a myriad of molecular mechanisms. The RAGE-ligand interactions play important roles in maintaining latent chronic inflammation, and oxidative damage underlying various pathological conditions like metabolic syndrome (MetS), neurodegenerative diseases, stroke, cardiovascular disorders, pulmonary disorders, cancer and infections. RAGE is thoroughly explored in knockout animals and human trials, targeted by small molecule inhibitors, peptides, diet, and natural compounds. But it is yet to be incorporated in the mainstream management of any ailment. This review performs an appraisal of the pathological mechanisms influenced by RAGE to uncover its prospects as a biomarker while also assessing its power to become a promising therapeutic target.
Collapse
Affiliation(s)
- Sinjini Sarkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS Deemed-to-be-University, V.L. Mehta Road, Vile Parle (W), Mumbai, 400056, India.
| |
Collapse
|
8
|
Tiwari H, Singh S, Sharma S, Gupta P, Verma A, Chattopadhaya A, Kumar B, Agarwal S, Kumar R, Gupta SK, Gautam V. Deciphering the landscape of triple negative breast cancer from microenvironment dynamics and molecular insights to biomarker analysis and therapeutic modalities. Med Res Rev 2024. [PMID: 39445844 DOI: 10.1002/med.22090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/05/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
Triple negative breast cancer (TNBC) displays a notable challenge in clinical oncology due to its invasive nature which is attributed to the absence of progesterone receptor (PR), estrogen receptor (ER), and human epidermal growth factor receptor (HER-2). The heterogenous tumor microenvironment (TME) of TNBC is composed of diverse constituents that intricately interact to evade immune response and facilitate cancer progression and metastasis. Based on molecular gene expression, TNBC is classified into four molecular subtypes: basal-like (BL1 and BL2), luminal androgen receptor (LAR), immunomodulatory (IM), and mesenchymal. TNBC is an aggressive histological variant with adverse prognosis and poor therapeutic response. The lack of response in most of the TNBC patients could be attributed to the heterogeneity of the disease, highlighting the need for more effective treatments and reliable prognostic biomarkers. Targeting certain signaling pathways and their components has emerged as a promising therapeutic strategy for improving patient outcomes. In this review, we have summarized the interactions among various components of the dynamic TME in TNBC and discussed the classification of its molecular subtypes. Moreover, the purpose of this review is to compile and provide an overview of the most recent data about recently discovered novel TNBC biomarkers and targeted therapeutics that have proven successful in treating metastatic TNBC. The emergence of novel therapeutic strategies such as chemoimmunotherapy, chimeric antigen receptor (CAR)-T cells-based immunotherapy, phytometabolites-mediated natural therapy, photodynamic and photothermal approaches have made a significant positive impact and have paved the way for more effective interventions.
Collapse
Affiliation(s)
- Harshita Tiwari
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Swati Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Sonal Sharma
- Department of General Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Priyamvada Gupta
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ashish Verma
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Amrit Chattopadhaya
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Brijesh Kumar
- Department of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Sakshi Agarwal
- Department of Obstetrics and Gynaecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Sanjeev Kumar Gupta
- Department of General Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Vibhav Gautam
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
9
|
Li Z, Yao L, Saravanakumar K, Thuy NTT, Kim Y, Xue C, Zheng X, Cho N. Lespedeza bicolor root extract exerts anti-TNBC potential by regulating FAK-related signalling pathways. Am J Cancer Res 2024; 14:4265-4285. [PMID: 39417178 PMCID: PMC11477838 DOI: 10.62347/mypg4066] [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: 07/14/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Lespedeza bicolor is a shrub plant that has been widely distributed in East Asia. The methanol extract from its LBR has been shown to exhibit anticancer and anti-bacterial effects. However, its anticancer efficacy in TNBC remains uncertain. This work aimed to study the anti-TNBC effect of LBR ethanol extract and its underlying mechanism. LBR triggered the cell death in TNBC through inhibiting cell proliferation, S-phase cell arrest, and induction of apoptosis. RNA-seq analysis revealed that the genes altered by LBR treatment were predominantly enriched in the cell adhesion. Notably, LBR inhibited phosphorylation and distribution of FAK. Furthermore, LBR demonstrated significant anticancer activity in xenograft tumors in mice through inhibiting cancer cell growth and inducing apoptosis. This work demonstrated the anticancer efficiency of LBR in TNBC without causing significant adverse effect, which providing a foundation for developing LBR based chemotherapeutic agents for breast cancer therapy.
Collapse
Affiliation(s)
- Zijun Li
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National UniversityGwangju 61186, Korea
| | - Lulu Yao
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National UniversityGwangju 61186, Korea
| | - Kandasamy Saravanakumar
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National UniversityGwangju 61186, Korea
| | - Nguyen Thi Thanh Thuy
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National UniversityGwangju 61186, Korea
| | - Yunyeong Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National UniversityGwangju 61186, Korea
| | - Chang Xue
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National UniversityGwangju 61186, Korea
| | - Xiaohui Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University1210 University Town, Wenzhou 325035, Zhejiang, China
| | - Namki Cho
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National UniversityGwangju 61186, Korea
| |
Collapse
|
10
|
Seki H, Kitabatake K, Tanuma SI, Tsukimoto M. Involvement of RAGE in radiation-induced acquisition of malignant phenotypes in human glioblastoma cells. Biochim Biophys Acta Gen Subj 2024; 1868:130650. [PMID: 38830560 DOI: 10.1016/j.bbagen.2024.130650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/23/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
Glioblastoma (GBM), a highly aggressive malignant tumor of the central nervous system, is mainly treated with radiotherapy. However, since irradiation may lead to the acquisition of migration ability by cancer cells, thereby promoting tumor metastasis and invasion, it is important to understand the mechanism of cell migration enhancement in order to prevent recurrence of GBM. The receptor for advanced glycation end products (RAGE) is a pattern recognition receptor activated by high mobility group box 1 (HMGB1). In this study, we found that RAGE plays a role in the enhancement of cell migration by γ-irradiation in human GBM A172 cells. γ-Irradiation induced actin remodeling, a marker of motility acquisition, and enhancement of cell migration in A172 cells. Both phenotypes were suppressed by specific inhibitors of RAGE (FPS-ZM1 and TTP488) or by knockdown of RAGE. The HMGB1 inhibitor ethyl pyruvate similarly suppressed γ-irradiation-induced enhancement of cell migration. In addition, γ-irradiation-induced phosphorylation of STAT3 was suppressed by RAGE inhibitors, and a STAT3 inhibitor suppressed γ-irradiation-induced enhancement of cell migration, indicating that STAT3 is involved in the migration enhancement downstream of RAGE. Our results suggest that HMGB1-RAGE-STAT3 signaling is involved in radiation-induced enhancement of GBM cell migration, and may contribute to GBM recurrence by promoting metastasis and invasion.
Collapse
Affiliation(s)
- Hiromu Seki
- Department of Radiation Biosciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Kazuki Kitabatake
- Department of Radiation Biosciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Sei-Ichi Tanuma
- Meikai University Research Institute of Odontology, Sakado, Saitama, Japan; Faculty of Human Science, University of Human Arts and Sciences, Iwatsuki, Saitama, Japan
| | - Mitsutoshi Tsukimoto
- Department of Radiation Biosciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan.
| |
Collapse
|
11
|
Shamsi A, Shahwan M, Anwar S, Ashames A, Khan MS, Yadav DK. Understanding the interactions between repurposed drugs sertindole and temoporfin with receptor for advanced glycation endproducts: Therapeutic implications in cancer and metabolic diseases. J Mol Model 2024; 30:170. [PMID: 38753123 DOI: 10.1007/s00894-024-05967-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 05/07/2024] [Indexed: 06/07/2024]
Abstract
CONTEXT In the pursuit of novel therapeutic possibilities, repurposing existing drugs has gained prominence as an efficient strategy. The findings from our study highlight the potential of repurposed drugs as promising candidates against receptor for advanced glycation endproducts (RAGE) that offer therapeutic implications in cancer, neurodegenerative conditions and metabolic syndromes. Through careful analyses of binding affinities and interaction patterns, we identified a few promising candidates, ultimately focusing on sertindole and temoporfin. These candidates exhibited exceptional binding affinities, efficacy, and specificity within the RAGE binding pocket. Notably, they displayed a pronounced propensity to interact with the active site of RAGE. Our investigation further revealed that sertindole and temoporfin possess desirable pharmacological properties that highlighted them as attractive candidates for targeted drug development. Overall, our integrated computational approach provides a comprehensive understanding of the interactions between repurposed drugs, sertindole and temoporfin and RAGE that pave the way for future experimental validation and drug development endeavors. METHODS We present an integrated approach utilizing molecular docking and extensive molecular dynamics (MD) simulations to evaluate the potential of FDA-approved drugs, sourced from DrugBank, against RAGE. To gain deeper insights into the binding mechanisms of the elucidated candidate repurposed drugs, sertindole and temoporfin with RAGE, we conducted extensive all-atom MD simulations, spanning 500 nanoseconds (ns). These simulations elucidated the conformational dynamics and stability of the RAGE-sertindole and RAGE-temoporfin complexes.
Collapse
Affiliation(s)
- Anas Shamsi
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| | - Moyad Shahwan
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, P.O. Box 346, Ajman, United Arab Emirates
| | - Saleha Anwar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Akram Ashames
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, P.O. Box 346, Ajman, United Arab Emirates
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Dharmendra Kumar Yadav
- Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, Incheon, Republic of Korea
| |
Collapse
|
12
|
Tang Q, Buonfiglio F, Böhm EW, Zhang L, Pfeiffer N, Korb CA, Gericke A. Diabetic Retinopathy: New Treatment Approaches Targeting Redox and Immune Mechanisms. Antioxidants (Basel) 2024; 13:594. [PMID: 38790699 PMCID: PMC11117924 DOI: 10.3390/antiox13050594] [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: 03/30/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Diabetic retinopathy (DR) represents a severe complication of diabetes mellitus, characterized by irreversible visual impairment resulting from microvascular abnormalities. Since the global prevalence of diabetes continues to escalate, DR has emerged as a prominent area of research interest. The development and progression of DR encompass a complex interplay of pathological and physiological mechanisms, such as high glucose-induced oxidative stress, immune responses, vascular endothelial dysfunction, as well as damage to retinal neurons. Recent years have unveiled the involvement of genomic and epigenetic factors in the formation of DR mechanisms. At present, extensive research explores the potential of biomarkers such as cytokines, molecular and cell therapies, antioxidant interventions, and gene therapy for DR treatment. Notably, certain drugs, such as anti-VEGF agents, antioxidants, inhibitors of inflammatory responses, and protein kinase C (PKC)-β inhibitors, have demonstrated promising outcomes in clinical trials. Within this context, this review article aims to introduce the recent molecular research on DR and highlight the current progress in the field, with a particular focus on the emerging and experimental treatment strategies targeting the immune and redox signaling pathways.
Collapse
Affiliation(s)
- Qi Tang
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (F.B.); (E.W.B.); (L.Z.); (N.P.); (C.A.K.)
| | | | | | | | | | | | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (F.B.); (E.W.B.); (L.Z.); (N.P.); (C.A.K.)
| |
Collapse
|
13
|
Chang TY, Lan KC, Wu CH, Sheu ML, Yang RS, Liu SH. Nε-(1-Carboxymethyl)-L-lysine/RAGE Signaling Drives Metastasis and Cancer Stemness through ERK/NFκB axis in Osteosarcoma. Int J Biol Sci 2024; 20:880-896. [PMID: 38250151 PMCID: PMC10797696 DOI: 10.7150/ijbs.90817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Osteosarcoma is an extremely aggressive bone cancer with poor prognosis. Nε-(1-Carboxymethyl)-L-lysine (CML), an advanced glycation end product (AGE), can link to cancer progression, tumorigenesis and metastasis, although the underlying mechanism remains unclear. The role of CML in osteosarcoma progression is still unclear. We hypothesized that CML could promote migration, invasion, and stemness in osteosarcoma cells. CML and its receptor (RAGE; receptor for AGE) were higher expressed at advanced stages in human osteosarcoma tissues. In mouse models, which streptozotocin was administered to induce CML accumulation in the body, the subcutaneous tumor growth was not affected, but the tumor metastasis using tail vein injection model was enhanced. In cell models (MG63 and U2OS cells), CML enhanced tumor sphere formation and acquisition of cancer stem cell characteristics, induced migration and invasion abilities, as well as triggered the epithelial-mesenchymal transition process, which were associated with RAGE expression and activation of downstream signaling pathways, especially the ERK/NFκB pathway. RAGE inhibition elicited CML-induced cell migration, invasion, and stemness through RAGE-mediated ERK/NFκB pathway. These results revealed a crucial role for CML in driving stemness and metastasis in osteosarcoma. These findings uncover a potential CML/RAGE connection and mechanism to osteosarcoma progression and set the stage for further investigation.
Collapse
Affiliation(s)
- Ting-Yu Chang
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuo-Cheng Lan
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Hung Wu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Rong-Sen Yang
- Department of Orthopedics, National Taiwan University Hospital, Taipei, Taiwan
| | - Shing-Hwa Liu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Pediatrics, College of Medicine, National Taiwan University & Hospital, Taipei, Taiwan
| |
Collapse
|
14
|
Reddy VP. Oxidative Stress in Health and Disease. Biomedicines 2023; 11:2925. [PMID: 38001926 PMCID: PMC10669448 DOI: 10.3390/biomedicines11112925] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Oxidative stress, resulting from the excessive intracellular accumulation of reactive oxygen species (ROS), reactive nitrogen species (RNS), and other free radical species, contributes to the onset and progression of various diseases, including diabetes, obesity, diabetic nephropathy, diabetic neuropathy, and neurological diseases, such as Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Parkinson's disease (PD). Oxidative stress is also implicated in cardiovascular disease and cancer. Exacerbated oxidative stress leads to the accelerated formation of advanced glycation end products (AGEs), a complex mixture of crosslinked proteins and protein modifications. Relatively high levels of AGEs are generated in diabetes, obesity, AD, and other I neurological diseases. AGEs such as Ne-carboxymethyllysine (CML) serve as markers for disease progression. AGEs, through interaction with receptors for advanced glycation end products (RAGE), initiate a cascade of deleterious signaling events to form inflammatory cytokines, and thereby further exacerbate oxidative stress in a vicious cycle. AGE inhibitors, AGE breakers, and RAGE inhibitors are therefore potential therapeutic agents for multiple diseases, including diabetes and AD. The complexity of the AGEs and the lack of well-established mechanisms for AGE formation are largely responsible for the lack of effective therapeutics targeting oxidative stress and AGE-related diseases. This review addresses the role of oxidative stress in the pathogenesis of AGE-related chronic diseases, including diabetes and neurological disorders, and recent progress in the development of therapeutics based on antioxidants, AGE breakers and RAGE inhibitors. Furthermore, this review outlines therapeutic strategies based on single-atom nanozymes that attenuate oxidative stress through the sequestering of reactive oxygen species (ROS) and reactive nitrogen species (RNS).
Collapse
Affiliation(s)
- V Prakash Reddy
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO 65409, USA
| |
Collapse
|