1
|
Zhang Z, Zhang Q, Wang Y. CAF-mediated tumor vascularization: From mechanistic insights to targeted therapies. Cell Signal 2025; 132:111827. [PMID: 40288665 DOI: 10.1016/j.cellsig.2025.111827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are a major component of the tumor microenvironment (TME) and play a crucial role in tumor progression. The biological properties of tumors, such as drug resistance, vascularization, immunosuppression, and metastasis are closely associated with CAFs. During tumor development, CAFs contribute to tumor progression by remodeling the extracellular matrix (ECM), inhibiting immune cell function, promoting angiogenesis, and facilitating tumor cell growth, invasion, and metastasis. Studies have shown that CAFs can promote endothelial cell proliferation by directly secreting cytokines such as vascular endothelial growth factor (VEGF) and fibroblast Growth Factor (FGF), as well as through exosomes. CAFs also secrete the chemokine stromal cell-derived factor 1 (SDF-1) to recruit endothelial progenitor cells (EPCs) into the peripheral blood and guide their migration to the tumor periphery. Additionally, CAFs can induce tumor cells to transform into "endothelial cells" that participate in vascular wall formation. However, the precise mechanisms remain to be further investigated. Due to their widespread presence in various solid tumors and their tumor-promoting function, CAFs are emerging as therapeutic targets. In this review, we summarize the specific mechanisms through which CAFs promote angiogenesis and outline current therapeutic strategies targeting CAF-induced vascularization, ongoing clinical trials targeting CAFs, and discuss potential future treatment approaches. We hope this will contribute to the advancement of CAF-targeted tumor treatment strategies.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Qing Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Yang Wang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
2
|
Birgersson M, Holm M, Gallardo-Dodd CJ, Chen B, Stepanauskaitė L, Hases L, Kutter C, Archer A, Williams C. Intestinal estrogen receptor beta modulates the murine colon tumor immune microenvironment. Cancer Lett 2025; 622:217661. [PMID: 40120798 DOI: 10.1016/j.canlet.2025.217661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Chronic inflammation contributes to the development of colorectal cancer, partly through its regulation of the microenvironment and antitumor immunity. Interestingly, women have a lower incidence of colorectal cancer, and estrogen treatment has been shown to reduce the occurrence of colorectal tumors. While intestinal estrogen receptor beta (ERβ, Esr2) can protect against colitis and colitis-induced cancer in mice, its role in shaping the tumor microenvironment remains unknown. In this study, we performed RNA sequencing to analyze the transcriptome of colonic epithelia and tumors from azoxymethane/dextran sulfate sodium-treated wild-type and intestinal ERβ knockout (ERβKOVil) mice and vehicle-treated controls. This revealed significant differences in gene expression and enriched biological processes influenced by sex and genotype, with immune-related responses being overrepresented. Deconvolution supported differential immune cell abundance and immunostaining showed that tumors from ERβKOVil mice displayed significantly increased macrophage infiltration, decreased T cell infiltration, and impaired natural killer cell infiltration. Further, ERβ mRNA levels in clinical colorectal tumors correlated with immune signaling profiles and better survival. Our findings indicate that intestinal ERβ promotes an antitumor microenvironment and could potentially affect the effectiveness of immunotherapy. These insights highlight the importance of ERβ in modulating antitumor immunity and underscore its therapeutic potential in colorectal cancer.
Collapse
Affiliation(s)
- Madeleine Birgersson
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Matilda Holm
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Carlos J Gallardo-Dodd
- Department of Microbiology, Tumor and Cell Biology, SciLifeLab, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Baizhen Chen
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden
| | - Lina Stepanauskaitė
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Linnea Hases
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Claudia Kutter
- Department of Microbiology, Tumor and Cell Biology, SciLifeLab, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Amena Archer
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Cecilia Williams
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83, Huddinge, Sweden.
| |
Collapse
|
3
|
Issa H, Singh L, Lai KS, Parusheva-Borsitzky T, Ansari S. Dynamics of inflammatory signals within the tumor microenvironment. World J Exp Med 2025; 15:102285. [DOI: 10.5493/wjem.v15.i2.102285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/31/2024] [Accepted: 01/11/2025] [Indexed: 04/16/2025] Open
Abstract
Tumor stroma, or tumor microenvironment (TME), has been in the spotlight during recent years for its role in tumor development, growth, and metastasis. It consists of a myriad of elements, including tumor-associated macrophages, cancer-associated fibroblasts, a deregulated extracellular matrix, endothelial cells, and vascular vessels. The release of proinflammatory molecules, due to the inflamed microenvironment, such as cytokines and chemokines is found to play a pivotal role in progression of cancer and response to therapy. This review discusses the major key players and important chemical inflammatory signals released in the TME. Furthermore, the latest breakthroughs in cytokine-mediated crosstalk between immune cells and cancer cells have been highlighted. In addition, recent updates on alterations in cytokine signaling between chronic inflammation and malignant TME have also been reviewed.
Collapse
Affiliation(s)
- Hala Issa
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Lokjan Singh
- Department of Microbiology, Karnali Academy of Health Sciences, Jumla 21200, Karnali, Nepal
| | - Kok-Song Lai
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Tina Parusheva-Borsitzky
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Shamshul Ansari
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| |
Collapse
|
4
|
Zhang H, Xu X, Li S, Huang H, Zhang K, Li W, Wang X, Yang J, Yin X, Qu C, Ni J, Dong X. Advances in nanoplatform-based multimodal combination therapy activating STING pathway for enhanced anti-tumor immunotherapy. Colloids Surf B Biointerfaces 2025; 250:114573. [PMID: 39983453 DOI: 10.1016/j.colsurfb.2025.114573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/24/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
Activation of the cyclic GMP-AMP synthase(cGAS)-stimulator of interferon genes (STING) has great potential to promote antitumor immunity. As a major effector of the cell to sense and respond to the aberrant presence of cytoplasmic double-stranded DNA (dsDNA), inducing the expression and secretion of type I interferons (IFN) and STING, cGAS-STING signaling pathway establishes an effective natural immune response, which is one of the fundamental mechanisms of host defense in organisms. In addition to the release of heterologous DNA due to pathogen invasion and replication, mitochondrial damage and massive cell death can also cause abnormal leakage of the body's own dsDNA, which is then recognized by the DNA receptor cGAS and activates the cGAS-STING signaling pathway. However, small molecule STING agonists suffer from rapid excretion, low bioavailability, non-specificity and adverse effects, which limits their therapeutic efficacy and in vivo application. Various types of nano-delivery systems, on the other hand, make use of the different unique structures and surface modifications of nanoparticles to circumvent the defects of small molecule STING agonists such as fast metabolism and low bioavailability. Also, the nanoparticles are precisely directed to the focal site, with their own appropriate particle size combined with the characteristics of passive or active targeting. Herein, combined with the cGAS-STING pathway to activate the immune system and kill tumor tissues directly or indirectly, which help maximize the use of the functions of chemotherapy, photothermal therapy(PTT), chemodynamic therapy(CDT), and radiotherapy(RT). In this review, we will discuss the mechanism of action of the cGAS-STING pathway and introduce nanoparticle-mediated tumor combination therapy based on the STING pathway. Collectively, the effective multimodal nanoplatform, which can activate cGAS-STING pathway for enhanced anti-tumor immunotherapy, has promising avenue clinical applications for cancer treatment.
Collapse
Affiliation(s)
- Huizhong Zhang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaohan Xu
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shiman Li
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Huating Huang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Ke Zhang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wenjing Li
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xinzhu Wang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jingwen Yang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xingbin Yin
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Changhai Qu
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jian Ni
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xiaoxv Dong
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
5
|
Wang N, Zhao TY, Ma X. Increased colorectal cancer risk in prediabetes: A meta-analysis. World J Diabetes 2025; 16:103403. [DOI: 10.4239/wjd.v16.i5.103403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/21/2025] [Accepted: 02/21/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Previous research yielded conflicting results regarding the association between prediabetes and colorectal cancer (CRC).
AIM To systematically assess the incidence of CRC in individuals with prediabetes compared with individuals with normoglycemia via a meta-analysis.
METHODS Relevant cohort studies were acquired by searching MEDLINE, Web of Science, and EMBASE. A random-effects model was applied to combine the findings after accounting for heterogeneity. Several subgroup analyses were conducted to assess the impact of study characteristics on the results.
RESULTS Eleven cohort studies involving 4996352 participants, including 383917 (7.7%) with prediabetes at baseline, were analyzed in this meta-analysis. Over a mean follow-up period of 6.5 years, the combined findings revealed that individuals with prediabetes at baseline had a higher likelihood of developing CRC than those with normoglycemia [risk ratio (RR) = 1.18, 95% confidence interval = 1.11 to 1.25, P < 0.001] with low statistical heterogeneity (I2 = 27%). Subgroup analyses indicated that the association between prediabetes and an increased risk of CRC was mainly observed in studies defining prediabetes using impaired fasting glucose (RR = 1.24) and slightly elevated hemoglobin A1c levels (RR = 1.18) but not in those that defined prediabetes using impaired glucose tolerance (RR = 1.06). Other study characteristics such as design, country, participant age and sex, the duration of follow-up, or adjustment for body mass index did not significantly impact the results (all P > 0.05).
CONCLUSION People with prediabetes might have a higher likelihood of developing CRC than individuals with normoglycemia.
Collapse
Affiliation(s)
- Na Wang
- Physical Examination Center, China-Japan Friendship Hospital, Beijing 100029, China
| | - Tian-Yi Zhao
- Physical Examination Center, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xiao Ma
- Physical Examination Center, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
6
|
Chaudhary P, Singha B, Abdel-Hafiz HA, Velegraki M, Sundi D, Satturwar S, Parwani AV, Grivennikov SI, You S, Goodridge HS, Ma Q, Chang Y, Ma A, Zheng B, Theodorescu D, Li Z, Li X. Sex differences in bladder cancer: understanding biological and clinical implications. Biol Sex Differ 2025; 16:31. [PMID: 40361239 PMCID: PMC12070554 DOI: 10.1186/s13293-025-00715-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Bladder cancer (BC) remains a significant global health concern, with substantial sex and racial disparities in incidence, progression, and outcomes. BC is the sixth most common cancer among males and the seventeenth most common among females worldwide. Over 90% of BC cases are urothelial carcinoma (UC) with high degrees of pathological heterogeneity. Molecular subtyping of BC has also revealed distinct luminal, basal, and neuroendocrine subtypes, each with unique genetic and immune signatures. Emerging research uncovers the biasing effects of the sex hormones with androgens increasing BC risk through both tumor cell intrinsic and extrinsic mechanisms. The sex chromosomes, including both the X and Y chromosomes, also contribute to the sex differences in BC. The effect of sex chromosome is both independent from and synergistic with the effects of sex hormones. Loss of the Y chromosome is frequently observed in BC patients, while an extra copy of the X chromosome confers better protection against BC in females than in males. Advent of advanced technologies such as multiomics and artificial intelligence will likely further improve the understanding of sex differences in BC, which may ultimately lead to personalized preventative and treatment strategies depending on the biological sex of patients. This review delves into the impacts of biology of sex on BC, emphasizing the importance of further research into sex-specific biology to improve cancer prevention and care.
Collapse
Affiliation(s)
- Prakash Chaudhary
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Biplab Singha
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hany A Abdel-Hafiz
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maria Velegraki
- Pelotonia Institute for Immuno‑Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Debasish Sundi
- Department of Urology, Division of Urologic Oncology, The Ohio State University, Comprehensive Cancer Center Board of Governors, Columbus, OH, USA
| | - Swati Satturwar
- Department of Pathology, Wexner Medical Center at The Ohio State University, Columbus, OH, 43210, USA
| | - Anil V Parwani
- Department of Pathology, Wexner Medical Center at The Ohio State University, Columbus, OH, 43210, USA
| | - Sergei I Grivennikov
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sungyong You
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Helen S Goodridge
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Yuzhou Chang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Anjun Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Bin Zheng
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dan Theodorescu
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Zihai Li
- Pelotonia Institute for Immuno‑Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Xue Li
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
7
|
Yarahmadi HB, Shahryari K, Bozorgi M, Shirdel A, Mohamadi Z, Rooshenas N, Karim Nezhad H, Mobaraki H, Aryannejad M, Emdadi A, Khosravian Y, Shahidi Marnani SA, SadatRafiei SK, Asadi Anar M, Marashi A, Khosravi F, Khodaei M. Dietary inflammatory index and the risk of esophageal cancer: a systematic review and meta-analysis. BMC Cancer 2025; 25:826. [PMID: 40319274 PMCID: PMC12048919 DOI: 10.1186/s12885-025-14199-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 04/22/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND AND AIM It is well-recognized that inflammation is an adaptive pathophysiological response in many types of cancer. Research on nutrition's critical role in inflammation, a risk factor for all forms of cancer, is growing. The dietary inflammatory index (DII) was created lately to assess if a diet is pro- or anti-inflammatory in terms of inflammation. Indeed, several studies have demonstrated the correlation between DII and the risk of several cancer types. This meta-analysis set out to look into the relationship between DII and the different forms of esophageal cancer. METHOD PubMed, Cochrane library, Embase, Scopus, and Web of Science databases were searched up to May 2024 to retrieve relevant articles. RAYYAN intelligent tool for systematic reviews was incorporated for the screening of studies. Original articles written in English Studies that investigated the inflammatory index of diet in individuals who developed esophageal cancer were included in this study.STATA v18 software was used to conduct the meta-analysis. Egger's test for publication bias assessment was implemented. Newcastle Ottawa scale was used to evaluate the qualities of the included studies. A plot digitizer was used to extract digital data. RESULT A total of 13 studies were included in the systematic review, with 6 studies contributing to the meta-analysis, comprising 10,150 participants. The participants were categorized into high and low DII groups, with the low DII group (n = 3,403) serving as the reference. The meta-analysis demonstrated a significant association between high DII and increased risk of esophageal cancer. Specifically, individuals in the high DII group were 29% more likely to develop esophageal cancer, with a pooled odds ratio (OR) of 1.29 (95% Confidence Interval [CI]: 1.16-1.43), as calculated using a random-effects model. Moderate heterogeneity was observed (I2 > 50%). Egger's test indicated evidence of publication bias (p < 0.05). Subgroup and sensitivity analyses confirmed the robustness of this association across populations and study designs. CONCLUSION our study concludes that a higher level of DII is associated with a higher risk of esophageal cancer development. This study suggests that modifying inflammatory properties of dietary patterns can reduce the risk of incidence of esophageal cancer.
Collapse
Affiliation(s)
| | - Kianoush Shahryari
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Bozorgi
- Shahid Beheshti Hospital, Kashan University of Medical Sciences, Kashan, Iran
| | - Ahmadreza Shirdel
- student research committee, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Zhina Mohamadi
- Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Negar Rooshenas
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Helia Karim Nezhad
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Hesam Mobaraki
- Faculty of Medicine, İstanbul Yeniyuzyil University, Istanbul, Turkey
| | - Majid Aryannejad
- Dental School, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Anahita Emdadi
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Yekta Khosravian
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | | | | | - Mahsa Asadi Anar
- College of Medicine, University of Arizona, 1501 N Campbell Ave, Tucson, AZ, 85724, USA.
| | - Amir Marashi
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farbod Khosravi
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Khodaei
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Nejat Dehkordi A, Maddahi M, Vafa P, Ebrahimi N, Aref AR. Salivary biomarkers: a promising approach for predicting immunotherapy response in head and neck cancers. Clin Transl Oncol 2025; 27:1887-1920. [PMID: 39377974 DOI: 10.1007/s12094-024-03742-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/21/2024] [Indexed: 04/27/2025]
Abstract
Head and neck cancers, including cancers of the mouth, throat, voice box, salivary glands, and nose, are a significant global health issue. Radiotherapy and surgery are commonly used treatments. However, due to treatment resistance and disease recurrence, new approaches such as immunotherapy are being explored. Immune checkpoint inhibitors (ICIs) have shown promise, but patient responses vary, necessitating predictive markers to guide appropriate treatment selection. This study investigates the potential of non-invasive biomarkers found in saliva, oral rinses, and tumor-derived exosomes to predict ICI response in head and neck cancer patients. The tumor microenvironment significantly impacts immunotherapy efficacy. Oral biomarkers can provide valuable information on composition, such as immune cell presence and checkpoint expression. Elevated tumor mutation load is also associated with heightened immunogenicity and ICI responsiveness. Furthermore, the oral microbiota may influence treatment outcomes. Current research aims to identify predictive salivary biomarkers. Initial studies indicate that tumor-derived exosomes and miRNAs present in saliva could identify immunosuppressive pathways and predict ICI response. While tissue-based markers like PD-L1 have limitations, combining multiple oral fluid biomarkers could create a robust panel to guide treatment decisions and advance personalized immunotherapy for head and neck cancer patients.
Collapse
Affiliation(s)
| | - Moein Maddahi
- Faculty of Density, Yeditepe University, Istanbul, Turkey
| | - Parinaz Vafa
- Faculty of Density, Yeditepe University, Istanbul, Turkey
| | - Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Amir Reza Aref
- Mass General Cancer Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
9
|
Yang Y, Hu Z, Ye Y, Wu H, Sun W, Wang N. Association of aggregate index of systemic inflammation with increased all-cause and cardiovascular mortality in female cancer patients. Front Oncol 2025; 15:1552341. [PMID: 40365348 PMCID: PMC12069043 DOI: 10.3389/fonc.2025.1552341] [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: 01/02/2025] [Accepted: 04/04/2025] [Indexed: 05/15/2025] Open
Abstract
Background Cancer is a leading cause of death, especially among women, with cancers like breast, ovarian, and cervical cancer presenting unique diagnostic and treatment challenges. Systemic inflammation plays a significant role in cancer progression, affecting both tumor development and therapeutic outcomes. Despite the established link between inflammation and cancer, comprehensive studies on the prognostic value of the Aggregate Index of Systemic Inflammation (AISI) in female cancer patients are lacking. This study explores the association between AISI and mortality outcomes, including all-cause and cardiovascular mortality, in female cancer patients. Methods This study analyzes data from the NHANES database and Dandong Central Hospital. Kaplan-Meier survival curves and multivariable Cox proportional hazards regression analyses were used to assess the relationship between AISI and all-cause and cardiovascular mortality. Restricted cubic spline plots and subgroup analyses were applied to explore potential interactions. Results Elevated AISI levels were strongly associated with increased all-cause and cardiovascular mortality. Patients in the highest AISI quartile demonstrated significantly higher mortality risks compared to those in the lowest quartile. ROC curve analysis indicated superior predictive performance of AISI over SII. Restricted cubic spline plots revealed a linear relationship, with mortality risk notably increasing when AISI levels were elevated. Conclusion AISI is a robust predictor of all-cause and cardiovascular mortality in female cancer patients. Its ease of measurement and strong prognostic value make it a valuable tool for risk assessment and management in this population.
Collapse
Affiliation(s)
- Ying Yang
- Dalian Medical University, Dalian, China
- The Dandong Central Hospital, Dandong, China
| | - Zelin Hu
- The Second Hospital of Dalian Medical University, Dalian, China
| | - Yuqin Ye
- Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Haoqi Wu
- The Second Hospital of Dalian Medical University, Dalian, China
| | - Wei Sun
- The Dandong Central Hospital, Dandong, China
| | - Ning Wang
- The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
10
|
Khalili-Tanha G, Radisky ES, Radisky DC, Shoari A. Matrix metalloproteinase-driven epithelial-mesenchymal transition: implications in health and disease. J Transl Med 2025; 23:436. [PMID: 40217300 PMCID: PMC11992850 DOI: 10.1186/s12967-025-06447-w] [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: 02/15/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a process in which epithelial cells, defined by apical-basal polarity and tight intercellular junctions, acquire migratory and invasive properties characteristic of mesenchymal cells. Under normal conditions, EMT directs essential morphogenetic events in embryogenesis and supports tissue repair. When dysregulated, EMT contributes to pathological processes such as organ fibrosis, chronic inflammation, and cancer progression and metastasis. Matrix metalloproteinases (MMPs)-a family of zinc-dependent proteases that degrade structural components of the extracellular matrix-sit at the nexus of this transition by dismantling basement membranes, activating pro-EMT signaling pathways, and cleaving adhesion molecules. When normally regulated, MMPs promote balanced ECM turnover and support the cyclical remodeling necessary for proper development, wound healing, and tissue homeostasis. When abnormally regulated, MMPs drive excessive ECM turnover, thereby promoting EMT-related pathologies, including tumor progression and fibrotic disease. This review provides an integrated overview of the molecular mechanisms by which MMPs both initiate and sustain EMT under physiological and disease conditions. It discusses how MMPs can potentiate EMT through TGF-β and Wnt/β-catenin signaling, disrupt cell-cell junction proteins, and potentiate the action of hypoxia-inducible factors in the tumor microenvironment. It discusses how these pathologic processes remodel tissues during fibrosis, and fuel cancer cell invasion, metastasis, and resistance to therapy. Finally, the review explores emerging therapeutic strategies that selectively target MMPs and EMT, ranging from CRISPR/Cas-mediated interventions to engineered tissue inhibitors of metalloproteinases (TIMPs), and demonstrates how such approaches may suppress pathological EMT without compromising its indispensable roles in normal biology.
Collapse
Affiliation(s)
- Ghazaleh Khalili-Tanha
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Alireza Shoari
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
11
|
Lu J, Liu X, Ji X, Jiang Y, Zuo A, Guo Z, Yang S, Peng H, Sun F, Lu D. Predicting PD-L1 status in NSCLC patients using deep learning radiomics based on CT images. Sci Rep 2025; 15:12495. [PMID: 40216830 PMCID: PMC11992188 DOI: 10.1038/s41598-025-91575-y] [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: 08/14/2024] [Accepted: 02/21/2025] [Indexed: 04/14/2025] Open
Abstract
Radiomics refers to the utilization of automated or semi-automated techniques to extract and analyze numerous quantitative features from medical images, such as computerized tomography (CT) or magnetic resonance imaging (MRI) scans. This study aims to develop a deep learning radiomics (DLR)-based approach for predicting programmed death-ligand 1 (PD-L1) expression in patients with non-small cell lung cancer (NSCLC). Data from 352 NSCLC patients with known PD-L1 expression were collected, of which 48.29% (170/352) were tested positive for PD-L1 expression. Tumor regions of interest (ROI) were semi-automatically segmented based on CT images, and DL features were extracted using Residual Network 50. The least absolute shrinkage and selection operator (LASSO) algorithm was used for feature selection and dimensionality reduction. Seven algorithms were used to build models, and the most optimal ones were identified. A combined model integrating DLR with clinical data was also developed. The predictive performance of each model was evaluated using the area under the curve (AUC) of the receiver operating characteristic (ROC) curve analysis. The DLR model, based on CT images, demonstrated an AUC of 0.85 (95% confidence interval (CI), 0.82-0.88), sensitivity of 0.80 (0.74-0.85), and specificity of 0.73 (0.70-0.77) for predicting PD-L1 status. The integrated model exhibited superior performance, with an AUC of 0.91 (0.87-0.95), sensitivity of 0.85 (0.82-0.89), and specificity of 0.75 (0.72-0.80). Our findings indicate that the DLR model holds promise as a valuable tool for predicting the PD-L1 status in patients with NSCLC, which can greatly assist in clinical decision-making and the selection of personalized treatment strategies.
Collapse
Affiliation(s)
- Jiameng Lu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
- Faculty of Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau Special Administrative Region, People's Republic of China
| | - Xinyi Liu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
| | - Xiaoqing Ji
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, Shandong, China
| | - Yunxiu Jiang
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
| | - Anli Zuo
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
| | - Zihan Guo
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
| | - Shuran Yang
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
| | - Haiying Peng
- Department of Respiratory and Critical Care Medicine, The Second People's Hospital of Yibin City, 644002, Yibin, People's Republic of China
| | - Fei Sun
- Department of Respiratory and Critical Care Medicine, Jining No.1 People's Hospital, 272000, Jining, People's Republic of China
| | - Degan Lu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China.
| |
Collapse
|
12
|
Zeng A, Quan Y, Tao H, Dai Y, Song L, Zhao J. The Role of Tetrahydrocurcumin in Tumor and Neurodegenerative Diseases Through Anti-Inflammatory Effects. Int J Mol Sci 2025; 26:3561. [PMID: 40332041 PMCID: PMC12027286 DOI: 10.3390/ijms26083561] [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: 02/24/2025] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 05/08/2025] Open
Abstract
Tetrahydrocurcumin (THC), a curcumin derivative, shows potential in oncology and neurology. It regulates NF-κB, reduces inflammation, promotes cancer cell apoptosis, inhibits tumor angiogenesis, and enhances antioxidants, aiding in treating inflammation-related cancers. In neurology, THC's anti-inflammatory and antioxidant properties protect neurons, reduce neuroinflammation, and support autophagy for cellular debris clearance, with its blood-brain barrier penetration offering a neuroprotective edge. Research on THC's therapeutic application must focus on improving delivery and bioavailability and confirming its clinical safety and efficacy.
Collapse
Affiliation(s)
- Anqi Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China; (A.Z.); (Y.Q.); (Y.D.)
- Sichuan Institute for Translational Chinese Medicine, Chengdu 610041, China
| | - Yunyun Quan
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China; (A.Z.); (Y.Q.); (Y.D.)
| | - Hongxia Tao
- West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Ying Dai
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China; (A.Z.); (Y.Q.); (Y.D.)
- Sichuan Institute for Translational Chinese Medicine, Chengdu 610041, China
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Junning Zhao
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China; (A.Z.); (Y.Q.); (Y.D.)
- Sichuan Institute for Translational Chinese Medicine, Chengdu 610041, China
| |
Collapse
|
13
|
Kahk NM, Mohamed FEA, Abdelhakeem MM, Bakr RB. Optimization of pyrazole/1,2,4-triazole as dual EGFR/COX-2 inhibitors: Design, synthesis, anticancer potential, apoptosis induction and cell cycle analysis. Eur J Med Chem 2025; 287:117340. [PMID: 39914141 DOI: 10.1016/j.ejmech.2025.117340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 01/25/2025] [Accepted: 01/27/2025] [Indexed: 02/24/2025]
Abstract
A novel series of pyrazol-4-yl-1,2,4-triazole-3-thiol derivatives 14a-l was designed, prepared and characterized by many spectroscopic techniques. All the novel compounds were screened for their anti-proliferative activity towards breast cancer cell line (MCF-7), colon cancer cell line (HT-29) and lung cancer cell line (A-549) utilizing celecoxib, erlotinib and osimertinib as standards. Compounds 14b, 14g and 14k were the most active towards HT-29, MCF-7 and A-549 cell lines, sequentially with IC50 = 1.20-2.93 μM compared with celecoxib (IC50 = 16.47-41.45 μM), erlotinib (IC50 = 1.95-33.57 μM) and osimertinib (IC50 = 0.75-3.45 μM). These most active derivatives 14b, 14g and 14k were further investigated for their inhibitory potential against COX and EGFR enzymes. These compounds 14b, 14g and 14k suppressed COX-2 (IC50 = 0.560-4.692 μM), EGFRWT (IC50 = 0.121-0.423 μM) and EGFRT790M (IC50 = 0.076-0.764 μM) enzymes. Compounds 14b, 14g and 14k displayed apoptosis induction by up-regulating Bax and down-regulating Bcl-2 protein levels. Cell cycle analysis recorded that exposure of MFC-7 cells to compound 14g resulted in a significant increase in the percentage of cells at the G2/M to 39.15 % compared to the standard erlotinib (9.87 %). Docking study of the most potent candidates 14b, 14g and 14k within COX-2, EGFRWT and EGFRT790M active regions was conducted to suggest the binding mode of these compounds inside these target enzymes.
Collapse
Affiliation(s)
- Nesma M Kahk
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni- Suef, 62514, Egypt
| | - Fatma E A Mohamed
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni- Suef, 62514, Egypt
| | - Marwa M Abdelhakeem
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni- Suef, 62514, Egypt
| | - Rania B Bakr
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni- Suef, 62514, Egypt.
| |
Collapse
|
14
|
Chen K, Zhao Y, Jin T, Xu T, Zhu C, Shen H, Xu X, Jiang Y, Wang Y, Xue S. Serum Amyloid A1 Mediates Paclitaxel Resistance via MD2-Dependent Pathways in Triple-Negative Breast Cancer. Drug Dev Res 2025; 86:e70047. [PMID: 40012323 DOI: 10.1002/ddr.70047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 11/15/2024] [Accepted: 01/06/2025] [Indexed: 02/28/2025]
Abstract
Triple-negative breast cancer (TNBC) is difficult to treat due to the lack of clear therapeutic targets. Paclitaxel (PTX) is commonly used to treat TNBC, but drug resistance limits its effectiveness. Myeloid differentiation protein 2 (MD2) and serum amyloid A1 (SAA1) are involved in various diseases, including infections, inflammatory diseases, and cancer. We investigated their role in PTX resistance to identify potential anti-TNBC drugs. In this study, we investigated the changes of SAA1 in TNBC tissues and its role in PTX-induced TNBC cells. Our study revealed SAA1 expressed in the human TNBC subtype and TNBC cells. PTX and CIS induce SAA1 in TNBC cells, and PTX induces inflammatory response via SAA1 in TNBC cells. MD2 blockade increased the sensitivity of TNBC cells to PTX, which was related to the expression of SAA1 during PTX-caused damage of TNBC cells. In further research, SAA1 binds to MD2, promotes the combination of TLR4/MD2 and TLR4/MyD88, activates the NF-κB signaling pathway, and creates the inflammatory microenvironment for cancer cells. Our study reports for the first time that the PTX/SAA1/MD2 axis exists in the PTX-resistance process, which could be a potential treatment target of PTX-resistance.
Collapse
Affiliation(s)
- Kangmin Chen
- Department of Oncology, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanni Zhao
- Department of Oncology, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tianyang Jin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tingxin Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Conglun Zhu
- Department of Pathology, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Hui Shen
- Department of Pathology, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Xiaohong Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongsheng Jiang
- Department of Oncology, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Shihang Xue
- Department of Oncology, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| |
Collapse
|
15
|
Wang R, Wang Z, Zhang M, Zhong D, Zhou M. Application of photosensitive microalgae in targeted tumor therapy. Adv Drug Deliv Rev 2025; 219:115519. [PMID: 39955076 DOI: 10.1016/j.addr.2025.115519] [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: 11/27/2024] [Revised: 01/02/2025] [Accepted: 01/18/2025] [Indexed: 02/17/2025]
Abstract
Microalgae present a novel and multifaceted approach to cancer therapy by modulating the tumor-associated microbiome (TAM) and the tumor microenvironment (TME). Through their ability to restore gut microbiota balance, reduce inflammation, and enhance immune responses, microalgae contribute to improved cancer treatment outcomes. As photosynthetic microorganisms, microalgae exhibit inherent anti-tumor, antioxidant, and immune-regulating properties, making them valuable in photodynamic therapy and tumor imaging due to their capacity to generate reactive oxygen species. Additionally, microalgae serve as effective drug delivery vehicles, leveraging their biocompatibility and unique structural properties to target the TME more precisely. Microalgae-based microrobots further expand their therapeutic potential by autonomously navigating complex biological environments, offering a promising future for precision-targeted cancer treatments. We position microalgae as a multifunctional agent capable of modulating TAM, offering novel strategies to enhance TME and improve the efficacy of cancer therapies.
Collapse
Affiliation(s)
- Ruoxi Wang
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310029, China; Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China; Zhejiang University-Ordos City Etuoke Banner Joint Research Center, Haining 314400, China
| | - Zhouyue Wang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China
| | - Min Zhang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China
| | - Danni Zhong
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Min Zhou
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310029, China; Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China; Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China; Zhejiang University-Ordos City Etuoke Banner Joint Research Center, Haining 314400, China.
| |
Collapse
|
16
|
Cao Z, Li J, Hu W, Xu J, Zhao F, Wang Y, Qin S, Liu M, Wang P, Duan J, Zhou W, Ding Z, Tang S, Ma X, Wang L. Near-Infrared Imaging Agent ABSi-148 Alleviates CA IX-Mediated Hypoxic Fibrosis in Inflammation-Cancer Transition. Adv Healthc Mater 2025; 14:e2404935. [PMID: 40099420 DOI: 10.1002/adhm.202404935] [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: 12/09/2024] [Revised: 03/07/2025] [Indexed: 03/19/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a formidable challenge due to its late diagnosis and intrinsic treatment resistance, exacerbates by its development from chronic inflammation to cancer transition (ICT). Here, this investigation aims to develop and evaluate ABSi-148, a novel near-infrared (NIR) agent targeting hypoxic carbonic anhydrase IX (CA IX), for its potential applications in ICT imaging and even PDAC treatment. ABSi-148 is synthesized from 4-(2-Aminoethyl) benzene sulfonamide (ABS), a sulfonamide derivative, conjugating with MHI-148 dye with merits of exceptional NIR-emitting traits, high biocompatibility, and deep tissue penetration imaging capability. It selectively accumulates in CoCl2-induced pancreatic stellate cells and pancreatic cancer cells via binding with transmembrane CA IX in vitro. Meanwhile, ABSi-148 effectively visualizes the early pancreatic lesion, and its long-term administration inhibits the progression of hypoxia-related fibrosis involved in pancreatic intraepithelial neoplasias (PanINs), and even PDAC progression in vivo. Besides, ABSi-148 monitors treatment efficacy and localizes hypoxic tumor regions, enhancing survival in tamoxifen combined with caerulein-induced KPC mice. Overall, ABSi-148 emerges as a theranostic NIR agent for precise diagnosis and targeted therapy in ICT of PDAC, promising to alleviate tumor progression and enhancing outcomes.
Collapse
Affiliation(s)
- Zhi Cao
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jingmin Li
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
- Internal medicine department, Guangzhou women and children's Medical Center, Guangzhou, 510623, China
| | - Weibin Hu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
- Department of Nuclear Medicine, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 518037, China
| | - Jian Xu
- Stroke center, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510799, China
| | - Fengyun Zhao
- Cancer Research Institute of Zhongshan City, Zhongshan City People's Hospital, Zhongshan, 528403, China
| | - Yishu Wang
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Shuanglin Qin
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, 410218, China
| | - Ming Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Ping Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510230, China
| | - Jingwei Duan
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Wensheng Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Zhaowei Ding
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510230, China
| | - Shaohui Tang
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Xiaodong Ma
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Lisheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| |
Collapse
|
17
|
Al Amin M, Bouhenni H, Zehravi M, Sweilam SH, Durgawale TP, Qureshi MS, Durgapal S, Haque MA, Vodeti R, Urs D, Shatu MM, Rab SO, Doukani K, Emran TB. Natural compounds and programmed necrosis: pioneering a new frontier in cancer treatments. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04050-w. [PMID: 40137962 DOI: 10.1007/s00210-025-04050-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025]
Abstract
Programmed necrosis, a controlled cell death method that bypasses resistance mechanisms that render apoptosis ineffective, is a potential cancer treatment target. Due to their diverse biological activities and low side effects, natural products are being explored as modulators of programmed necrosis pathways. This review highlights the potential of natural compounds to target cancer cells while preserving healthy tissues and their interaction with essential programmed necrosis mechanisms like ferroptosis and necroptosis. Recent developments have identified various types of programmable necrosis, including necroptosis, ferroptosis, pyroptosis, proptosis, mitochondrial permeability transition-driven necrosis, and oncosis. Natural compounds are increasingly being utilized as a primary source of anti-cancer medications, providing new cancer treatments. This review demonstrates the molecular mechanisms behind lipid peroxidation, mixed lineage kinase domain-like protein, and receptor-interacting protein kinases (RIPK1 and RIPK3) inducing cell death. Recent research has identified natural compounds like polyphenols, alkaloids, and terpenoids that can modulate pathways and benefit preclinical cancer models. The review underscores the potential of natural compounds in developing innovative cancer treatments by integrating pharmacology and cellular signaling knowledge. Integrating natural compound studies and programmed necrosis research presents a promising avenue for oncologists to overcome treatment resistance. Natural compounds have shown potential in developing programmed necrosis as a novel cancer treatment approach, enhancing therapeutic effectiveness and minimizing side effects through preclinical research, pharmacology, and molecular biology.
Collapse
Affiliation(s)
- Md Al Amin
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, 1216, Bangladesh.
| | - Hasna Bouhenni
- Laboratory of Agrobiotechnology and Nutrition in Semi-Arid Zones, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah, 51418, Saudi Arabia.
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo, 11829, Egypt
| | - Trupti Pratik Durgawale
- Department of Pharmaceutical Chemistry, Krishna Institute of Pharmacy Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, Maharashtra, India
| | - Mohammad Shamim Qureshi
- Department of Pharmacognosy & Phytochemistry, Anwarul Uloom College of Pharmacy, New Mallepally, Hyderabad, 500001, India
| | - Sumit Durgapal
- Department of Pharmaceutics, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Premnagar, Dehradun, Uttarakhand, 248007, India
| | - M Akiful Haque
- School of Pharmacy, Anurag University, Venkatapur, Hyderabad, Telangana , 500088, India
| | - Rajeshwar Vodeti
- Deportment of Pharmaceutics, School of Pharmacy, Anurag University, Hyderabad, India
| | - Deepadarshan Urs
- Inflammation Research Laboratory, Department of Studies & Research in Biochemistry, Mangalore University, Jnana Kaveri Post Graduate Centre, Kodagu, Karnataka, 571232, India
| | - Mst Maharunnasa Shatu
- Department of Botany, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Koula Doukani
- Laboratory of Agrobiotechnology and Nutrition in Semi-Arid Zones, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
- Laboratory of Animal Production Sciences and Techniques, Faculty of Nature and Life Sciences, University of Abdelhamid Ibn Badis, Mostaganem, Algeria
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, 1216, Bangladesh.
| |
Collapse
|
18
|
Su Y, Bai Q, Zhang W, Xu B, Hu T. The Role of Long Non-Coding RNAs in Modulating the Immune Microenvironment of Triple-Negative Breast Cancer: Mechanistic Insights and Therapeutic Potential. Biomolecules 2025; 15:454. [PMID: 40149989 PMCID: PMC11939868 DOI: 10.3390/biom15030454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly heterogeneous and aggressive subtype of breast cancer that faces therapeutic challenges due to a shortage of effective targeted therapies. The complex biology of TNBC renders its clinical management fraught with difficulties, especially regarding the immune microenvironment of the tumor. In recent years, long non-coding RNAs (lncRNAs) have been recognized as important gene regulators with key roles in tumor development and microenvironmental regulation. Previous studies have shown that lncRNAs play important roles in the immune microenvironment of TNBC, including the regulation of tumor immune escape and the function of tumor-infiltrating immune cells. However, despite the increasing research on lncRNAs, there are still many unanswered questions, such as their specific mechanism of action and how to effectively utilize them as therapeutic targets. Therefore, the aim of this study was to review the mechanisms of lncRNAs in the TNBC immune microenvironment, explore their regulatory roles in tumor immune escape and immune cell infiltration, and explore their prospects as potential therapeutic targets. By integrating the latest research results, this study aims to provide new ideas and directions for future TNBC treatment.
Collapse
Affiliation(s)
- Yongcheng Su
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (Q.B.); (W.Z.)
| | - Qingquan Bai
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (Q.B.); (W.Z.)
| | - Wenqing Zhang
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (Q.B.); (W.Z.)
| | - Beibei Xu
- Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Tianhui Hu
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (Q.B.); (W.Z.)
- Shenzhen Research Institute, Xiamen University, Shenzhen 518057, China
| |
Collapse
|
19
|
Siedlecka-Kroplewska K, Kmiec Z, Zmijewski MA. The Interplay Between Autophagy and Apoptosis in the Mechanisms of Action of Stilbenes in Cancer Cells. Antioxidants (Basel) 2025; 14:339. [PMID: 40227400 PMCID: PMC11939748 DOI: 10.3390/antiox14030339] [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: 01/31/2025] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
Plant-based stilbenes are low-molecular-weight polyphenolic compounds that exhibit anti-oxidant, anti-microbial, anti-fungal, anti-inflammatory, anti-diabetic, cardioprotective, neuroprotective, and anti-cancer activities. They are phytoalexins produced in diverse plant species in response to stress, such as fungal and bacterial infections or excessive UV irradiation. Plant-derived dietary products containing stilbenes are common components of the human diet. Stilbenes appear to be promising chemopreventive and chemotherapeutic agents. Accumulating evidence indicates that stilbenes are able to trigger both apoptotic and autophagic molecular pathways in many human cancer cell lines. Of note, the molecular crosstalk between autophagy and apoptosis under cellular stress conditions determines the cell fate. The autophagy and apoptosis relationship is complex and depends on the cellular context, e.g., cell type and cellular stress level. Apoptosis is a type of regulated cell death, whereas autophagy may act as a pro-survival or pro-death mechanism depending on the context. The interplay between autophagy and apoptosis may have an important impact on chemotherapy efficiency. This review focuses on the in vitro effects of stilbenes in different human cancer cell lines concerning the interplay between autophagy and apoptosis.
Collapse
Affiliation(s)
| | - Zbigniew Kmiec
- Department of Anatomy and Histology, School of Medicine, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland;
| | | |
Collapse
|
20
|
Chavda VP, Bezbaruah R, Ahmed N, Alom S, Bhattacharjee B, Nalla LV, Rynjah D, Gadanec LK, Apostolopoulos V. Proinflammatory Cytokines in Chronic Respiratory Diseases and Their Management. Cells 2025; 14:400. [PMID: 40136649 PMCID: PMC11941495 DOI: 10.3390/cells14060400] [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: 11/22/2024] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Pulmonary homeostasis can be agitated either by external environmental insults or endogenous factors produced during respiratory/pulmonary diseases. The lungs counter these insults by initiating mechanisms of inflammation as a localized, non-specific first-line defense response. Cytokines are small signaling glycoprotein molecules that control the immune response. They are formed by numerous categories of cell types and induce the movement, growth, differentiation, and death of cells. During respiratory diseases, multiple proinflammatory cytokines play a crucial role in orchestrating chronic inflammation and structural changes in the respiratory tract by recruiting inflammatory cells and maintaining the release of growth factors to maintain inflammation. The issue aggravates when the inflammatory response is exaggerated and/or cytokine production becomes dysregulated. In such instances, unresolving and chronic inflammatory reactions and cytokine production accelerate airway remodeling and maladaptive outcomes. Pro-inflammatory cytokines generate these deleterious consequences through interactions with receptors, which in turn initiate a signal in the cell, triggering a response. The cytokine profile and inflammatory cascade seen in different pulmonary diseases vary and have become fundamental targets for advancement in new therapeutic strategies for lung diseases. There are considerable therapeutic approaches that target cytokine-mediated inflammation in pulmonary diseases; however, blocking specific cytokines may not contribute to clinical benefit. Alternatively, broad-spectrum anti-inflammatory approaches are more likely to be clinically effective. Herein, this comprehensive review of the literature identifies various cytokines (e.g., interleukins, chemokines, and growth factors) involved in pulmonary inflammation and the pathogenesis of respiratory diseases (e.g., asthma, chronic obstructive pulmonary, lung cancer, pneumonia, and pulmonary fibrosis) and investigates targeted therapeutic treatment approaches.
Collapse
Affiliation(s)
- Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India; (R.B.); (N.A.); (S.A.)
- Institute of Pharmacy, Assam Medical College and Hospital, Dibrugarh 786002, Assam, India
| | - Nasima Ahmed
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India; (R.B.); (N.A.); (S.A.)
| | - Shahnaz Alom
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India; (R.B.); (N.A.); (S.A.)
- Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India; (B.B.); (D.R.)
| | - Bedanta Bhattacharjee
- Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India; (B.B.); (D.R.)
| | - Lakshmi Vineela Nalla
- Department of Pharmacology, GITAM School of Pharmacy, GITAM (Deemed to be University), Rushikonda, Visakhapatnam 530045, Andhra Pradesh, India;
| | - Damanbhalang Rynjah
- Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India; (B.B.); (D.R.)
| | - Laura Kate Gadanec
- Institute for Health and Sport, Immunology and Translational Research Group, Victoria University, Werribee, VIC 3030, Australia;
| | - Vasso Apostolopoulos
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia;
| |
Collapse
|
21
|
Li J, Ren Y, Li H, Zheng Z. Rac1 overexpression promotes Treg-derived cytokines to mediate choroidal neovascularization in wet age-related macular degeneration. Braz J Med Biol Res 2025; 58:e14187. [PMID: 40053038 PMCID: PMC11884777 DOI: 10.1590/1414-431x2024e14187] [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: 08/14/2024] [Accepted: 12/12/2024] [Indexed: 03/10/2025] Open
Abstract
Age-related macular degeneration (AMD), particularly the wet form characterized by choroidal neovascularization, is a leading cause of vision loss. Dysregulation of regulatory T cells (Tregs), key modulators of inflammatory responses, may contribute to wet AMD pathogenesis. This study explored the involvement of Tregs and the Rac1 signaling pathway in modulating Treg-derived cytokine expression and their role in choroidal neovascularization during wet AMD progression. Peripheral blood samples from healthy controls, dry AMD patients, and wet AMD patients were collected. An in vitro transmembrane co-culture system of Tregs and human choroidal endothelial cells (HCECs) was employed to investigate the impact of Tregs (with or without Rac1 silencing) on the angiogenic phenotype of HCECs. A mouse model of AMD was established to evaluate the effects of a Rac1 inhibitor and IL-10/TGF-β neutralization on Tregs and choroidal neovascularization. An increased Treg percentage in the CD4+ T lymphocyte population was found in the peripheral blood samples of wet AMD patients. Tregs from wet AMD patients showed an increased expression of Rac1 and an elevated production of IL-10 and TGF-β1. Rac1 silencing suppressed Treg stability and differentiation, and impaired the pro-angiogenic effect of Tregs on HCECs. In the animal model of AMD, the administration of a Rac1 inhibitor or neutralizing antibodies against IL-10/TGF-β1 reduced Treg abundance and attenuated choroidal neovascularization. Rac1 upregulation in Tregs promoted IL-10 and TGF-β1 production to mediate choroidal neovascularization in wet AMD. Targeting Rac1 and Treg-derived IL-10/TGF-β1 production in Tregs may serve as a strategy to ameliorate AMD progression.
Collapse
Affiliation(s)
- Juanjuan Li
- Affiliated Hospital of Yunnan University (Second People's Hospital of Yunnan Province, Yunnan Eye Hospital), Kunming, Yunnan, China
| | - Yuling Ren
- Affiliated Hospital of Yunnan University (Second People's Hospital of Yunnan Province, Yunnan Eye Hospital), Kunming, Yunnan, China
| | - Hua Li
- Affiliated Hospital of Yunnan University (Second People's Hospital of Yunnan Province, Yunnan Eye Hospital), Kunming, Yunnan, China
| | - Zhikun Zheng
- Affiliated Hospital of Yunnan University (Second People's Hospital of Yunnan Province, Yunnan Eye Hospital), Kunming, Yunnan, China
| |
Collapse
|
22
|
Mu M, Inoue H, Mao D, Sougawa N, Goda S. β1 Integrin/FAK signaling regulates interleukin-8 production in human gingival epithelial Ca9-22 cells. J Oral Biosci 2025; 67:100615. [PMID: 39826873 DOI: 10.1016/j.job.2025.100615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVES Interleukin-8 (IL-8), a proinflammatory factor in human tissues, plays an important role in inflammation. Type IV collagen, a key component of the basement membrane, interacts with integrins, which are primary receptors in the extracellular matrix (ECM). Integrins are essential for the regulation of various cellular behaviors and signal transduction pathways. However, the relationship between type IV collagen, β1 integrin, and gingival epithelial cells is poorly understood. The aim in this study was to elucidate the effect of the interaction between type IV collagen and β1 integrin on IL-8 secretion in human gingival epithelial cells (Ca9-22). METHODS Ca9-22 cells were treated with or without type IV collagen, and IL-8 production was assessed using an enzyme-linked immunosorbent assay (ELISA). The role of β1 integrin was investigated using a β1 integrin-neutralizing antibody. Western blotting was performed to measure the phosphorylation levels of the relevant proteins. The effects of the focal adhesion kinase (FAK) inhibitor Y15 and the MEK inhibitor U0126 on β1 integrin/FAK and Erk1/2 MAPK pathways in IL-8 production were evaluated to explore the involvement of these signaling pathways. RESULTS β1 integrin induced IL-8 secretion in the Ca9-22 cells by regulating FAK, Erk1/2, and p130Cas proteins. p130Cas was independent of FAK, whereas Erk1/2 functioned downstream of FAK. Inhibition of FAK or Erk1/2 substantially reduced IL-8 secretion, highlighting their pivotal roles in this signaling pathway. CONCLUSION β1 integrin promotes IL-8 secretion in Ca9-22 cells via the β1 integrin/FAK/Erk1/2 signaling pathway. These findings elucidate the pathogenesis of periodontitis and provide a foundation for the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Meili Mu
- Graduate School of Dentistry, Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Hiroshi Inoue
- Department of Physiology, Osaka Dental University, Osaka, Japan.
| | - Dan Mao
- Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Nagako Sougawa
- Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Seiji Goda
- Department of Physiology, Osaka Dental University, Osaka, Japan.
| |
Collapse
|
23
|
Benedik NS, Proj M, Steinebach C, Sova M, Sosič I. Targeting TAK1: Evolution of inhibitors, challenges, and future directions. Pharmacol Ther 2025; 267:108810. [PMID: 39909209 DOI: 10.1016/j.pharmthera.2025.108810] [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: 11/15/2024] [Revised: 01/07/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
The increasing incidence of inflammatory and malignant diseases signifies the need to develop first-in-class drugs with novel mechanisms of action. In this respect, the transforming growth factor (TGF)-β-activated kinase 1 (TAK1), an essential part of several signaling pathways, is considered relevant and promising. This manuscript provides a brief overview of the signal transduction orchestrated by TAK1 within these pathways, followed by an in-depth and thorough analysis of the chemical matter demonstrated to inhibit this kinase. Special attention is given to the selectivity profiling of inhibitors, as well as to the outcomes of their biological characterization. Because published TAK1 inhibitors differ significantly in their kinome selectivity, active-site binding, and biological activity, we hope that this review will allow a judicial estimation of their quality and usefulness for TAK1-addressing assays. Our thoughts on the perspectives and possible developments of the field are also provided to assist scientists who are involved in the design and development of TAK1-targeting modulators.
Collapse
Affiliation(s)
- Nika Strašek Benedik
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000 Ljubljana, Slovenia
| | - Matic Proj
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000 Ljubljana, Slovenia
| | - Christian Steinebach
- Pharmaceutical Institute, Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Matej Sova
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000 Ljubljana, Slovenia
| | - Izidor Sosič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
24
|
Zhang W, Xu C. Exosomal SphK1 from colorectal cancer cells promotes cancer cell migration and activates hepatic stellate cells. Mol Med Rep 2025; 31:73. [PMID: 39930826 PMCID: PMC11795250 DOI: 10.3892/mmr.2025.13438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/03/2025] [Indexed: 02/14/2025] Open
Abstract
Exosomes are small extracellular vesicles that are naturally released into body fluids by cells. They are rich in bioactive molecules such as proteins. Sphingosine kinase 1 (SphK1) is an important potential drug target for the treatment of cancer due to its functions to regulate cancer cell migration, growth, apoptosis and angiogenesis. Tumor exosomes abundantly surround primary tumors, exchanging and transferring information between cells and modulating cancer progression. Given the importance of exosomes, the involvement of exosomal SphK1 from colorectal cancer (CRC) cells in the migration of these cells and the activation of hepatic stellate cells was investigated. Firstly, the plasma exosomal SphK1 protein expression, tested by ELISA, was compared between patients with CRC without metastasis and those with liver metastasis. The results revealed that plasma exosomal SphK1 levels were significantly upregulated in patients with liver metastasis of CRC. Secondly, exosomes with different expression levels of SphK1, which were regulated by cell transfection, were isolated from CRC cells to evaluate their effect on the expression levels of E‑cadherin and vimentin in these cells, as assessed by western blotting. The results demonstrated that depletion of exosomal SphK1 partially reversed the exosome‑induced migration of CRC cells, and caused decreased vimentin and increased E‑cadherin levels. Thirdly, the effects of exosomes from CRC cells, with different expression levels of SphK1, on hepatic stellate cell activation were investigated, with α‑SMA, TNF‑α and TGF‑β levels assessed by western blotting in LX‑2 cells. Moreover, AKT and phosphorylated (p‑)AKT levels were also assessed by western blotting. The results revealed that exosomes activated hepatic stellate cells by upregulating p‑AKT, and depletion of exosomal SphK1 partially reversed this effect. Furthermore, the application of an AKT agonist reversed the inhibition of hepatic stellate cell activation, which was induced by the depletion of exosomal SphK1. Finally, investigation of cell viability, analyzed by CCK‑8 assay, and assessment of PCNA as a proliferation marker, analyzed by western blot, revealed that the culture supernatant of the activated hepatic stellate cells promoted the viability of CRC cells. Overall, these results demonstrated that exosomal SphK1 increased the migration of CRC cells, and activated hepatic stellate cells by regulating p‑AKT. This suggests that exosomal SphK1 may serve a key role in the migration of CRC cells and potentially the liver metastasis of CRC.
Collapse
Affiliation(s)
- Wenlu Zhang
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Chunyan Xu
- Department of Gastroenterology and Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong 250100, P.R. China
| |
Collapse
|
25
|
Yagci T, Genc S, Dundar R, Altiner HI, Taghizadehghalehjoughi A. A Combination of Anatolian Propolis and Curcumin Protects Fibroblasts Against Beclomethasone (Nazal Steroid)-Induced Oxidative Stress by Modulating IL-25, MMP-2, VEGF, and FGF-2 Expressions. Pharmaceuticals (Basel) 2025; 18:326. [PMID: 40143104 PMCID: PMC11946772 DOI: 10.3390/ph18030326] [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: 01/08/2025] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Nasal steroids are commonly prescribed in ear, nose, and throat clinics. It is observed that the use of nasal steroids is increasing due to the prevalence of allergic rhinitis. Because beclomethasone (BCM) toxicity is low, it is highly preferred in allergic rhinitis. The rate of toxicity increases with the increase in the duration and dose of BCM use. However, the protective mechanism of Anatolian propolis (AP) and curcumin (Cur) against BCM toxicity has not been fully explained. Aim: The study evaluates the potential BCM-induced toxicity effect on VEGF, MMP-2, IL-25, and IL-10 parameters after Cur and AP treatment. Materials and Methods: Cell viability, oxidative stress, and gene expression were used for toxicity evaluation. Results: AP 2.5 mg/mL and Cur 16 µg/mL show high viability and antioxidant capacity. BCM increased the levels of IL-25, IL-10, and MMP-2, and a decrease was detected in the expression levels of FGF-2 and VEGF. Conclusions: AP and Cur show effective healing, and AP has been shown to improve inflammation more effectively than Cur. However, the combination of AP and Cur significantly improved the induced toxicity effects.
Collapse
Affiliation(s)
- Tarik Yagci
- Department of Otorhinolaryngology, Faculty of Medicine, Şeyh Edebali University, 11230 Bilecik, Turkey; (T.Y.); (R.D.); (H.I.A.)
| | - Sidika Genc
- Department of Medical Pharmacology, Faculty of Medicine, Şeyh Edebali University, 11230 Bilecik, Turkey;
| | - Riza Dundar
- Department of Otorhinolaryngology, Faculty of Medicine, Şeyh Edebali University, 11230 Bilecik, Turkey; (T.Y.); (R.D.); (H.I.A.)
| | - Halil Ibrahim Altiner
- Department of Otorhinolaryngology, Faculty of Medicine, Şeyh Edebali University, 11230 Bilecik, Turkey; (T.Y.); (R.D.); (H.I.A.)
| | - Ali Taghizadehghalehjoughi
- Department of Medical Pharmacology, Faculty of Medicine, Şeyh Edebali University, 11230 Bilecik, Turkey;
| |
Collapse
|
26
|
D’Amico RC, Nagashima S, Carstens LB, Bertoldi KDG, Mataruco S, Honório D’Agostini JC, Hlatchuk EC, da Silva SB, de Noronha L, Baena CP. COVID-19 Induces Greater NLRP3 Inflammasome Activation in Obese Patients than Other Chronic Illnesses: A Case-Control Study. Int J Mol Sci 2025; 26:1541. [PMID: 40004007 PMCID: PMC11855377 DOI: 10.3390/ijms26041541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Obesity has been identified as an independent risk factor for severe COVID-19 unfavorable outcomes. Several factors, such as increased ACE2 receptor expression and chronic inflammation, can contribute to this relationship, yet the activation of the NLRP3 inflammasome pathway is also a key element. Our primary goal was to determine whether chronic NLRP3 inflammasome activation in people with obesity is different in critical COVID-19 and in critical chronic conditions. A retrospective analysis was conducted using clinical data and post-mortem lung tissue samples from 14 COVID-19 patients with obesity (group A) and 9 patients with obesity who died from non-COVID-19 causes (group B). Immunohistochemical analysis assessed twelve markers related to the NLRP3 inflammasome pathway. Group A showed a significantly higher expression of ASC (p = 0.0387) and CASP-1 (p = 0.0142). No significant differences were found for IL-8, TNF-α, NF-kB, NLRP3, IL-1β, and gasdermin-D. Group B had higher levels of IL-6 (p < 0.0001), IL-18 (p = 0.002), CASP-9 (p < 0.0001), and HIF (p = 0.0327). We concluded that COVID-19 activates the NLRP3 inflammasome pathway, possibly leading to pyroptotic cell death mediated by caspase-1. In contrast, people with obesity without COVID-19, despite exhibiting some markers of the NLRP3 inflammasome, are more likely to experience necroptosis mediated by caspase-9.
Collapse
Affiliation(s)
- Raíssa Campos D’Amico
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | - Seigo Nagashima
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
| | - Lucas Baena Carstens
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | - Karina de Guadalupe Bertoldi
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
| | - Sabrina Mataruco
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | | | - Elisa Carolina Hlatchuk
- School of Medicine, Universidade Federal do Paraná, Curitiba 80060-240, Paraná, Brazil; (J.C.H.D.); (E.C.H.)
| | - Sofia Brunoro da Silva
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | - Lucia de Noronha
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
- School of Medicine, Universidade Federal do Paraná, Curitiba 80060-240, Paraná, Brazil; (J.C.H.D.); (E.C.H.)
| | - Cristina Pellegrino Baena
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| |
Collapse
|
27
|
Kaltsas A, Giannakas T, Stavropoulos M, Kratiras Z, Chrisofos M. Oxidative Stress in Benign Prostatic Hyperplasia: Mechanisms, Clinical Relevance and Therapeutic Perspectives. Diseases 2025; 13:53. [PMID: 39997060 PMCID: PMC11854834 DOI: 10.3390/diseases13020053] [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: 01/14/2025] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Benign prostatic hyperplasia (BPH) is among the most common conditions affecting men as they age, resulting in lower urinary tract symptoms (LUTS) that can profoundly impact quality of life. While historically attributed primarily to androgenic imbalances, current evidence implicates additional factors-particularly oxidative stress (OS) and chronic inflammation-in BPH pathogenesis. This review aims to synthesize research on the interplay between OS, inflammation, and hormonal regulation in BPH, emphasizing their clinical relevance and potential therapeutic implications. METHODS A comprehensive review of peer-reviewed literature was conducted focusing on mechanistic studies, clinical trials, and observational reports. Searches included data on ROS generation, antioxidant capacity, inflammatory mediators, and their contribution to pathological prostatic overgrowth. Potential interventions targeting OS-such as antioxidant supplementation, anti-inflammatory drugs, vitamin D receptor agonists, and phytotherapeutics-were also evaluated for their efficacy and safety profiles. RESULTS Chronic inflammation and OS were consistently identified within hyperplastic prostate tissue. Excessive ROS production, diminished antioxidant defense, and sustained cytokine release create a proproliferative and antiapoptotic environment, accelerating disease progression. Metabolic comorbidities (e.g., obesity, insulin resistance) further exacerbate these imbalances. Standard therapies (α-blockers and 5-ARIs) effectively relieve symptoms but do not directly address the oxidative-inflammatory axis. Emerging evidence suggests that pharmacological and dietary approaches targeting OS and inflammation may reduce prostate volume expansion and alleviate LUTS. CONCLUSIONS Findings indicate that OS and inflammation are key contributors to BPH progression. Incorporating antioxidant and anti-inflammatory strategies alongside conventional treatments holds promise for improving clinical outcomes and patient quality of life. Future research should focus on validating OS-specific biomarkers and optimizing personalized therapy regimens.
Collapse
Affiliation(s)
| | | | | | | | - Michael Chrisofos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (T.G.); (M.S.); (Z.K.)
| |
Collapse
|
28
|
Verma R, Zeyaullah M, Singh V, Saxena PS, Koch B, Kumar M. Chitosan-Functionalized Fluorescent Calcium Carbonate Nanoparticle Loaded with Methotrexate: Future Theranostics for Triple Negative Breast Cancer. ACS Biomater Sci Eng 2025; 11:981-999. [PMID: 39883480 DOI: 10.1021/acsbiomaterials.4c02390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Herein, fluorescent calcium carbonate nanoclusters encapsulated with methotrexate (Mtx) and surface functionalized with chitosan (25 nm) (@Calmat) have been developed for the imaging and treatment of triple-negative breast cancer (TNBC). These biocompatible, pH-sensitive nanoparticles demonstrate significant potential for targeted therapy and diagnostic applications. The efficacy of nanoparticles (NPs) was evaluated in MDA-MB-231 TNBC cell lines. The enhanced permeability and retention effect facilitated the accumulation of NPs, in tumor-bearing rats, as confirmed by in vivo fluorescence imaging. Treatment with @Calmat resulted in a marked reduction in pro-inflammatory cytokines, with levels of IL-6 (1225 ± 67 pg/mL), IL-1β (379 ± 69 pg/mL), and TNF-α (14.1 ± 2 pg/mL), in contrast to the diseased control group (IL-6: 2223 ± 99; IL-1β: 1632 ± 90; TNF-α: 40 ± 3 pg/mL). A similar trend was observed for liver and kidney function biomarkers. Mechanistic studies revealed that @Calmat treatment activates the Bax/Bcl-2 signaling pathway, leading to cell cycle arrest in the G1 phase and subsequent late-phase apoptosis. As a result, the tumor inhibition rate reached 88%, with 80% of treated rats surviving beyond 100 days. These findings highlight the strong potential of @Calmat as a dual-function theranostic agent for the management of TNBC.
Collapse
Affiliation(s)
- Rinki Verma
- School of Biomedical Engineering, IIT (BHU), Varanasi 221005, India
| | - Md Zeyaullah
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Virendra Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Preeti Suman Saxena
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Biplob Koch
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Manoj Kumar
- Nano 2 Micro Material Design Lab, Department of Chemical Engineering and Technology, IIT (BHU), Varanasi 221005, India
| |
Collapse
|
29
|
Wu J, Deng Z, Lei X, Xu Z, Tan C, Tang Y, Sheng X, Yang N. Prognostic evaluation of non-muscle invasive bladder cancer with P-CRP and its nomogram. Front Oncol 2025; 15:1406585. [PMID: 39963109 PMCID: PMC11830596 DOI: 10.3389/fonc.2025.1406585] [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: 04/11/2024] [Accepted: 01/14/2025] [Indexed: 02/20/2025] Open
Abstract
Purpose To investigate the impact of the product of preoperative platelet count and C-reactive protein (P-CRP) on the postoperative prognosis of patients with non-muscle invasive bladder cancer (NMIBC), and to construct a Nomogram to predict the recurrence-free survival (RFS) of NMIBC patients based on pathological data. Methods A retrospective analysis was conducted on the clinical data of 164 NMIBC patients who underwent transurethral resection of bladder tumors (TURBT) at the Second Affiliated Hospital of University of South China from January 2013 to December 2019. The endpoint of the study was the RFS. Kaplan-Meier (KM) method and Cox regression were used for analysis to identify independent factors affecting RFS. Then, the Nomogram was used to visualize the results of the multivariate analysis that were statistically significant and related to the RFS of NMIBC patients. Finally, the predictive ability of the model was evaluated using the concordance index (C-index) and calibration curves. Results Before the end of the follow-up, the RFS was 88.3% at 1 year, 75.5% at 2 years, and 58.5% at 3 years. KM curves showed that P-CRP (HR=0.357, 95% CI: 0.204-0.625, P<0.001), number of tumors (HR=2.658, 95% CI: 1.572-4.494, P<0.001), tumor size (HR=2.271, 95% CI: 1.377-3.745, P=0.001), T stage of the tumor (HR=2.026, 95% CI: 1.233-3.329, P=0.005), and tumor G grade (G2: HR=1.615, 95% CI: 0.48-5.433, G3: HR=3.361, 95% CI: 1.022-11.054) were independent factors affecting the RFS of NMIBC patients after TURBT. The Nomogram could estimate the risk of tumor recurrence at 1, 2, and 3 years postoperatively. The Nomogram model incorporating P-CRP parameters had a higher predictive accuracy than the classic model that only included EORTC risk group parameters. Conclusion Preoperative P-CRP has a certain impact on the RFS of NMIBC patients after TURBT. The Nomogram incorporating P-CRP, number of tumors, tumor size, T stage, and tumor pathological grading can better predict the postoperative recurrence risk of NMIBC patients.
Collapse
Affiliation(s)
- Junyun Wu
- The Second Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Zhixuan Deng
- Institute of Cell Biology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xu Lei
- The Central Hospital of Shaoyang, Shaoyang, Hunan, China
| | - Zhiyao Xu
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Chenxi Tan
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yunqiao Tang
- The Second Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xi Sheng
- The Second Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Ning Yang
- The Second Affiliated Hospital of University of South China, Hengyang, Hunan, China
| |
Collapse
|
30
|
Qi L, Wang J, Hou S, Liu S, Zhang Q, Zhu S, Liu S, Zhang S. Unraveling the tumor microenvironment of esophageal squamous cell carcinoma through single-cell sequencing: A comprehensive review. Biochim Biophys Acta Rev Cancer 2025; 1880:189264. [PMID: 39805342 DOI: 10.1016/j.bbcan.2025.189264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a highly heterogeneous and aggressive malignancy. The progression, invasiveness, and metastatic potential of ESCC are shaped by a multitude of cells within the tumor microenvironment (TME), including tumor cells, immune cells, endothelial cells, as well as fibroblasts and other cell types. Recent advancements in single-cell sequencing technologies have significantly enhanced our comprehension of the diverse landscape of ESCC. Single-cell multi-omics technology, particularly single-cell transcriptome sequencing, have shed light on the expression profiles of individual cells and the molecular characteristics of distinct tumor cell populations. This review summarizes the latest literature on single-cell research in the field of ESCC, aiming to elucidate the heterogeneity of tumor cells, immune cells, and stromal cells at the single-cell level. Furthermore, it explores the impact of cellular interactions within the TME on the progression of ESCC. By compiling a comprehensive overview of single-cell omics research on ESCC, this article aims to enhance our understanding of ESCC diagnosis and treatment by elucidating the intricate interplay within the TME. It explores the cellular composition, spatial arrangement, and functional attributes of the ESCC TME, offering potential therapeutic targets and biomarkers for personalized treatment strategies.
Collapse
Affiliation(s)
- Lingyu Qi
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Jiaxin Wang
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Songyuan Hou
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Siying Liu
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Qian Zhang
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Shengtao Zhu
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Si Liu
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China.
| | - Shutian Zhang
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China.
| |
Collapse
|
31
|
Li C, Zhao Y, He C, Wang X, Ren Q, Gai X, Wang H. IL-17E facilitates cell proliferation and epithelial-mesenchymal transition in A549 NSCLC cells by regulating the NF-κB pathway. Pathol Res Pract 2025; 266:155792. [PMID: 39753017 DOI: 10.1016/j.prp.2024.155792] [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: 08/22/2024] [Accepted: 12/23/2024] [Indexed: 05/01/2025]
Abstract
OBJECTIVE Interleukin-17 E (IL-17E) is a pro-inflammatory cytokine that participates in the inflammatory response and tumorigenesis. However, the function of IL-17E in non-small cell lung cancer (NSCLC) remains largely unknown. METHODS The clinical value of IL-17E was determined by immunohistochemistry (IHC) in 75 cases of NSCLC tissues. Furthermore, A549 cells were added with recombinant human IL-17E (rhIL-17E) or transfected with IL-17E siRNAs to evaluate the impact on cell proliferation, apoptosis, and epithelial-mesenchymal transition (EMT), as well as explore the link between IL-17E and the NF-κB pathway. Experimental techniques include CCK-8, EdU, colony formation, RT-qPCR, western blotting, flow cytometry, wound-healing, transwell and immunofluorescence assay. RESULTS IL-17E levels was elevated in NSCLC tissues and cells, which was related to higher TNM staging, positive lymph node metastasis and decreased tumor differentiation degree. Exogenous recombinant human IL-17E (rhIL-17E) treatment promoted cell proliferation, reduced cell apoptosis, and increased the level of Bcl-2/BAX. Moreover, it enhanced cell migration, invasion, EMT and phosphorylation levels of NF-κB p65. Inversely, knocking down endogenous IL-17E in A549 cells had the opposite effect. Blocking the NF-κB pathway with BAY-117082 reduced IL-17E expression and reversed the malignant effects induced by IL-17E on A549 cells. CONCLUSION IL-17E facilitates NSCLC progression by promoting cell proliferation and EMT via the NF-κB pathway. IL-17E could serve as a potential strategy for NSCLC treatment.
Collapse
Affiliation(s)
- Chun Li
- Department of Immunology, School of Basic Medical Sciences, Beihua University, Jilin 132013, China
| | - Ying Zhao
- Department of Immunology, School of Basic Medical Sciences, Beihua University, Jilin 132013, China
| | - Chengyuan He
- Department of Immunology, School of Basic Medical Sciences, Beihua University, Jilin 132013, China
| | - Xingxiang Wang
- Department of Immunology, School of Basic Medical Sciences, Beihua University, Jilin 132013, China
| | - Qiaotong Ren
- Department of Immunology, School of Basic Medical Sciences, Beihua University, Jilin 132013, China
| | - Xiaodong Gai
- Department of Immunology, School of Basic Medical Sciences, Beihua University, Jilin 132013, China.
| | - Hefei Wang
- Department of Obstetrics and Gynaecology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
32
|
Yao Z, Shang W, Yang F, Tian W, Zhao G, Xu X, Md RZ, Tian T, Li W, Huang M, Zhao Y, Huang Q. Nomogram for predicting severe abdominal adhesions prior to definitive surgery in patients with anastomotic fistula post-small intestine resection: a cohort study. Int J Surg 2025; 111:2046-2054. [PMID: 39705136 DOI: 10.1097/js9.0000000000002191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 11/20/2024] [Indexed: 12/22/2024]
Abstract
BACKGROUND This study aimed to develop and validate a nomogram for predicting the presence of severe intra-abdominal adhesions before definitive surgery (DS) for anastomotic fistula following small intestine resection (SIR). METHODS Patients were enrolled from January 2009 to October 2023 and were randomly divided (2:1) into development and validation cohorts. Predictors of severe adhesion were identified and integrated into a nomogram. The nomogram's performance was evaluated through calibration, discrimination, and clinical utility. Results : A total of 414 patients were included, with 276 in the development cohort and 138 in the validation cohort. Severe adhesion was diagnosed in 54 (13%) patients, including 37 (13.4%) in the development cohort and 17 (12.3%) in the validation cohort ( P = 0.76). Five predictors were identified: Sequential Organ Failure Assessment score, duration of early-stage abdominal infection, preoperative albumin (Alb) <35 g/L, visceral to subcutaneous fat area ratio, and preoperative C-reactive protein >10 mg/L. The nomogram demonstrated robust discrimination, with a concordance index (C-index) of 0.80 (95% CI, 0.76-0.90) in internal validation, and was well-calibrated. In the validation cohort, the model maintained good discrimination (C-index = 0.79; 95% CI, 0.67-0.94) and calibration. Decision curve analysis affirmed the nomogram's clinical utility. CONCLUSION This study introduces a practical nomogram for assessing the risk of severe abdominal adhesion prior to DS in patients undergoing surgery for anastomotic fistula after SIR.
Collapse
Affiliation(s)
- Zheng Yao
- Department of General Surgery, Jiangning Hospital, Nanjing, Jiangsu, China
| | - Weiwei Shang
- Department of General Surgery, Jiangning Hospital, Nanjing, Jiangsu, China
| | - Fan Yang
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weiliang Tian
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guoping Zhao
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Xu
- Department of General Surgery, Jiangning Hospital, Nanjing, Jiangsu, China
| | - Risheng Zhao Md
- Department of General Surgery, Jiangning Hospital, Nanjing, Jiangsu, China
| | - Tao Tian
- Department of General Surgery, Shanghai 9th Hospital, Shanghai, China
| | - Wuhan Li
- Department of General Surgery, Anhui Provincial Hospital, Hefei, Anhui, China
| | - Ming Huang
- Department of General Surgery, Jiangning Hospital, Nanjing, Jiangsu, China
| | - Yunzhao Zhao
- Department of General Surgery, Jiangning Hospital, Nanjing, Jiangsu, China
| | - Qian Huang
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
33
|
Hosoda K, Shimizu A, Kubota K, Notake T, Kitagawa N, Yoshizawa T, Sakai H, Hayashi H, Yasukawa K, Soejima Y. Clinical significance of the Naples prognostic score in predicting short- and long-term postoperative outcomes of patients with hepatocellular carcinoma. World J Surg 2025; 49:502-511. [PMID: 39631788 PMCID: PMC11798678 DOI: 10.1002/wjs.12448] [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: 05/02/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND The Naples prognostic score (NPS) is a remarkable marker of short- and long-term outcomes in various types of cancer. However, its impact on the postoperative outcomes of hepatocellular carcinoma remains controversial. This study aimed to clarify the impact of the NPS on the prognosis and incidence of postoperative complications in hepatocellular carcinoma. METHODS Patients with hepatocellular carcinoma (n = 374) were categorized into high- and low-Naples prognostic score groups; their postoperative outcomes were compared. Prognostic and risk factors for severe postoperative complications were identified using multivariate analyses. RESULTS The low-Naples prognostic score group had significantly longer overall and recurrence-free survivals than the high-Naples prognostic score group (p = 0.03 and 0.04, respectively). Subgroup analysis revealed a superior predictive value of the NPS in the group with a single tumor (p = 0.03), tumor diameter ≤5 cm (p = 0.04), and tumor stage I or II (p = 0.04). A high NPS was an independent prognostic factor for overall survival (hazard ratio, 1.45; 95% confidence interval (CI), 1.01-2.05; and p = 0.04). The NPS 2-4 group had a higher incidence of the Clavien-Dindo grade ≥ IIIa postoperative complications than the 0-1 group (p = 0.03) and a score of 2-4 was identified as an independent risk factor for the Clavien-Dindo grade ≥ IIIa postoperative complications (odds ratio, 2.06; 95% CI, 1.01-4.20; and p = 0.05). CONCLUSIONS The NPS effectively predicts postoperative outcomes in patients with hepatocellular carcinoma.
Collapse
Affiliation(s)
- Kiyotaka Hosoda
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation, and Pediatric SurgeryDepartment of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Akira Shimizu
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation, and Pediatric SurgeryDepartment of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Koji Kubota
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation, and Pediatric SurgeryDepartment of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Tsuyoshi Notake
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation, and Pediatric SurgeryDepartment of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Noriyuki Kitagawa
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation, and Pediatric SurgeryDepartment of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Takahiro Yoshizawa
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation, and Pediatric SurgeryDepartment of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Hiroki Sakai
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation, and Pediatric SurgeryDepartment of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Hikaru Hayashi
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation, and Pediatric SurgeryDepartment of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Koya Yasukawa
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation, and Pediatric SurgeryDepartment of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Yuji Soejima
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation, and Pediatric SurgeryDepartment of SurgeryShinshu University School of MedicineMatsumotoJapan
| |
Collapse
|
34
|
Godos J, Currenti W, Ferri R, Lanza G, Caraci F, Frias-Toral E, Guglielmetti M, Ferraris C, Lipari V, Carvajal Altamiranda S, Galvano F, Castellano S, Grosso G. Chronotype and Cancer: Emerging Relation Between Chrononutrition and Oncology from Human Studies. Nutrients 2025; 17:529. [PMID: 39940387 PMCID: PMC11819666 DOI: 10.3390/nu17030529] [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: 12/30/2024] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
Fasting-feeding timing is a crucial pattern implicated in the regulation of daily circadian rhythms. The interplay between sleep and meal timing underscores the importance of maintaining circadian alignment in order to avoid creating a metabolic environment conducive to carcinogenesis following the molecular and systemic disruption of metabolic performance and immune function. The chronicity of such a condition may support the initiation and progression of cancer through a variety of mechanisms, including increased oxidative stress, immune suppression, and the activation of proliferative signaling pathways. This review aims to summarize current evidence from human studies and provide an overview of the potential mechanisms underscoring the role of chrononutrition (including time-restricted eating) on cancer risk. Current evidence shows that the morning chronotype, suggesting an alignment between physiological circadian rhythms and eating timing, is associated with a lower risk of cancer. Also, early time-restricted eating and prolonged nighttime fasting were also associated with a lower risk of cancer. The current evidence suggests that the chronotype influences cancer risk through cell cycle regulation, the modulation of metabolic pathways and inflammation, and gut microbiota fluctuations. In conclusion, although there are no clear guidelines on this matter, emerging evidence supports the hypothesis that the role of time-related eating (i.e., time/calorie-restricted feeding and intermittent/periodic fasting) could potentially lead to a reduced risk of cancer.
Collapse
Affiliation(s)
- Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (J.G.)
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, 95123 Catania, Italy
| | - Walter Currenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (J.G.)
| | | | - Giuseppe Lanza
- Oasi Research Institute-IRCCS, 94018 Troina, Italy
- Department of Surgery and Medical-Surgical Specialties, University of Catania, 95125 Catania, Italy
| | - Filippo Caraci
- Oasi Research Institute-IRCCS, 94018 Troina, Italy
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
| | - Evelyn Frias-Toral
- School of Medicine, Universidad Espíritu Santo, Samborondón 0901952, Ecuador
| | - Monica Guglielmetti
- Human Nutrition and Eating Disorder Research Center, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy
- Laboratory of Food Education and Sport Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy
| | - Cinzia Ferraris
- Human Nutrition and Eating Disorder Research Center, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy
- Laboratory of Food Education and Sport Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy
| | - Vivian Lipari
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain
- Universidad de La Romana, La Romana 22000, Dominican Republic
- Universidad Internacional Iberoamericana, Campeche 24560, Mexico
| | - Stefanía Carvajal Altamiranda
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain
- Universidade Internacional do Cuanza, Cuito EN250, Angola
- Fundación Universitaria Internacional de Colombia, Bogotá 111321, Colombia
| | - Fabio Galvano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (J.G.)
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, 95123 Catania, Italy
| | - Sabrina Castellano
- Department of Educational Sciences, University of Catania, 95124 Catania, Italy
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (J.G.)
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, 95123 Catania, Italy
| |
Collapse
|
35
|
Dahl-Wilkie H, Gomez J, Kelley A, Manjit K, Mansoor B, Kanumuri P, Pardo S, Molleur D, Falah R, Konakalla AR, Omiyale M, Weintraub S, Delk NA. Chronic IL-1-Exposed LNCaP Cells Evolve High Basal p62-KEAP1 Complex Accumulation and NRF2/KEAP1-Dependent and -Independent Hypersensitive Nutrient Deprivation Response. Cells 2025; 14:192. [PMID: 39936983 PMCID: PMC11816438 DOI: 10.3390/cells14030192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/25/2025] [Accepted: 01/26/2025] [Indexed: 02/13/2025] Open
Abstract
Chronic inflammation is a cancer hallmark and chronic exposure to interleukin-1 (IL-1) transforms castration-sensitive prostate cancer (PCa) cells into more fit castration-insensitive PCa cells. p62 is a scaffold protein that protects cells from nutrient deprivation via autophagy and from cytotoxic reactive oxygen via NFκB and NRF2 antioxidant signaling. Herein, we report that the LNCaP PCa cell line acquires high basal accumulation of the p62-KEAP1 complex when chronically exposed to IL-1. p62 promotes non-canonical NRF2 antioxidant signaling by binding and sequestering KEAP1 to the autophagosome for degradation. But despite high basal p62-KEAP1 accumulation, only two of several NRF2-induced genes analyzed, GCLC and HMOX1, showed high basal mRNA levels, suggesting that the high basal p62-KEAP1 accumulation does not result in overall high basal NRF2 activity. Nutrient starvation induces NRF2-dependent GCLC upregulation and HMOX1 repression, and we found that chronic IL-1-exposed LNCaP cells show hypersensitivity to serum starvation-induced GCLC and HMOX1 regulation. Thus, chronic IL-1 exposure affects cell response to nutrient stress. While HMOX1 expression remains NRF2/KEAP1-dependent in chronic IL-1-exposed LNCaP cells, GCLC expression is NRF2/KEAP1-independent. Furthermore, the high basal p62-KEAP1 complex accumulation is not required to regulate GCLC or HMOX1 expression, suggesting cells chronically exposed to IL-1 evolve a novel NRF2-independent role for the p62/KEAP1 axis.
Collapse
Affiliation(s)
- Haley Dahl-Wilkie
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Jessica Gomez
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Anastasia Kelley
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Kirti Manjit
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Basir Mansoor
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Preethi Kanumuri
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Sammy Pardo
- Department of Biochemistry & Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (S.P.); (D.M.); (S.W.)
| | - Dana Molleur
- Department of Biochemistry & Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (S.P.); (D.M.); (S.W.)
| | - Rafah Falah
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Anisha R. Konakalla
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Morolake Omiyale
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| | - Susan Weintraub
- Department of Biochemistry & Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (S.P.); (D.M.); (S.W.)
| | - Nikki A. Delk
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA; (H.D.-W.); (J.G.); (A.K.); (K.M.); (B.M.); (P.K.); (R.F.); (A.R.K.); (M.O.)
| |
Collapse
|
36
|
Niu ZH, Lin L, Peng HY, Zheng XZ, Wang MY, Sun FX, Xu CJ. The prognostic value of systemic inflammation response index in digestive system carcinomas: a systematic review and meta-analysis. BMC Gastroenterol 2025; 25:34. [PMID: 39856542 PMCID: PMC11761727 DOI: 10.1186/s12876-025-03635-2] [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: 08/19/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Digestive system carcinomas (DSC) constitute a significant proportion of solid tumors, with incidence rates rising steadily each year. The systemic inflammation response index (SIRI) has been identified as a potential prognostic marker for survival in various types DSC. This meta-analysis aimed to evaluate the prognostic value of SIRI in patients with DSC. METHODS We conducted a comprehensive literature search of PubMed, Web of Science Core Collection, Embase, and Cochrane Library databases, searching for studies published from inception to May 30, 2023. Eligible studies included cohort studies that assessed the association between pre-treatment SIRI levels and DSC prognosis. We extracted and synthesized hazard ratios (HRs) and 95% confidence intervals (CIs) using STATA/SE 12.0, stratifying HRs based on univariable and multivariable analysis. Due to substantial heterogeneity, we applied a random-effect model for all pooled analyses. The primary outcome of interest was the overall survival (OS), while secondary outcomes included progression-free survival (PFS), disease-free survival (DFS), time to progression (TTP), and disease specific survival (DSS). Publication bias was evaluated using Begg's test and Egger's tests. RESULTS A total of 34 cohort studies encompassing 9628 participants were included in this meta-analysis. Notable heterogeneity was observedin the OS (I2 = 76.5%, p < 0.001) and PFS (I2 = 82.8%, p = 0.001) subgroups, whereas no significant heterogeneity was detected in the DFS, TTP, and DSS subgroups. Elevated SIRI was found to be significantly associated with shorter OS (HR = 1.98, 95% CI: 1.70-2.30, tau2 = 0.0966) and poorer PFS (HR = 2.36, 95% CI: 1.58-3.53, tau2 = 0.1319), DFS (HR = 1.80, 95% CI: 1.61-2.01, tau2 < 0.0001), TTP (HR = 2.03, 95% CI: 1.47-2.81, tau2 = 0.0232), and DSS (HR = 1.99, 95% CI: 1.46-2.72, tau2 < 0.0001). Furthermore, an increase in SIRI following treatment was linked to reduced OS, TTP, and DFS, while a decrease in SIRI post-treatment corresponded with improved OS, TTP, and DFS compared to baseline levels. CONCLUSIONS Elevated SIRI is associated with poorer clinical outcomes in patients with DSC. This index may serve as a valuable prognostic biomarker, offering a promising tool for predicting survival in DSC patients. PROSPERO REGISTRATION NUMBER: CRD42023430962.
Collapse
Affiliation(s)
- Zuo-Hu Niu
- Department of Infections, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Li Lin
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Hong-Ye Peng
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xin-Zhuo Zheng
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Mi-Yuan Wang
- School of Management, Beijing University of Chinese medicine, Beijing, China
| | - Feng-Xia Sun
- Department of Infections, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Chun-Jun Xu
- Department of Infections, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| |
Collapse
|
37
|
Sheng F, Li M, Yu JM, Yang SY, Zou L, Yang GJ, Zhang LL. IL-33/ST2 axis in diverse diseases: regulatory mechanisms and therapeutic potential. Front Immunol 2025; 16:1533335. [PMID: 39925809 PMCID: PMC11802536 DOI: 10.3389/fimmu.2025.1533335] [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: 11/23/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
Interleukin-33 (IL-33) is a nuclear factor and member of the IL-1 cytokine family. IL-33 is mainly expressed by epithelial and endothelial cells and exerts its function through interaction with various immune cells, and binding to its receptor can form the IL-33/Suppression of tumorigenicity 2 (ST2) signaling pathway. While most cytokines are actively synthesized within cells, IL-33 is produced passively in response to tissue damage or cell necrosis, indicating its role as a signaling molecule following cellular infection, stress, or trauma. IL-33/ST2 signaling pathway has been proved to play diverse role in the pathological process of central nervous system disorders, cancer, fibrosis, autoimmune diseases, etc. Although research on the IL-33/ST2 signaling pathway has deepened recently, relevant treatment strategies have been proposed, and even targeted drugs are in the preclinical stage; further research on the effect of the IL-33/ST2 signaling pathway in different diseases is still necessary, to provide a clearer understanding of the different roles of IL-33/ST2 in disease progression and to develop new drugs and treatment strategies. Because IL-33/ST2 plays an important role in the occurrence and progression of diseases, the study of therapeutic drugs targeting this pathway is also necessary. This review focused on recent studies on the positive or negative role of IL-33/ST2 in different diseases, as well as the current related drugs targeting IL-33/ST2 in the preclinical and clinical stage. The mechanism of IL-33/ST2 in different diseases and its mediating effect on different immune cells have been summarized, as well as the antibody drugs targeting IL-33 or ST2, natural compounds with a mediating effect, and small molecule substances targeting relative pathway. We aim to provide new ideas and treatment strategies for IL-33/ST2-related drugs to treat different diseases.
Collapse
Affiliation(s)
- Feiya Sheng
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Mi Li
- College of Pharmacy, Chengdu University, Chengdu, China
| | - Jia-Mei Yu
- College of Pharmacy, Chengdu University, Chengdu, China
| | - Si-Yu Yang
- College of Pharmacy, Chengdu University, Chengdu, China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Chengdu University, Chengdu, China
| | - Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro−Products, Ningbo University, Ningbo, China
| | - Le-Le Zhang
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China
| |
Collapse
|
38
|
Ortega MA, Boaru DL, De Leon-Oliva D, De Castro-Martinez P, Minaya-Bravo AM, Casanova-Martín C, Barrena-Blázquez S, Garcia-Montero C, Fraile-Martinez O, Lopez-Gonzalez L, Saez MA, Alvarez-Mon M, Diaz-Pedrero R. The Impact of Klotho in Cancer: From Development and Progression to Therapeutic Potential. Genes (Basel) 2025; 16:128. [PMID: 40004457 PMCID: PMC11854833 DOI: 10.3390/genes16020128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Klotho, initially identified as an anti-aging gene, has been shown to play significant roles in cancer biology. Alongside α-Klotho, the β-Klotho and γ-Klotho isoforms have also been studied; these studies showed that Klotho functions as a potential tumor suppressor in many different cancers by inhibiting cancer cell proliferation, inducing apoptosis and modulating critical signaling pathways such as the Wnt/β-catenin and PI3K/Akt pathways. In cancers such as breast cancer, colorectal cancer, hepatocellular carcinoma, ovarian cancer, and renal cell carcinoma, reduced Klotho expression often correlates with a poor prognosis. In addition, Klotho's role in enhancing chemotherapy sensitivity and its epigenetic regulation further underscores its potential as a target for cancer treatments. This review details Klotho's multifaceted contributions to cancer suppression and its potential as a therapeutic target, enhancing the understanding of its significance in cancer treatment and prognoses.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Ana M. Minaya-Bravo
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Carlos Casanova-Martín
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Silvestra Barrena-Blázquez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Department of General and Digestive Surgery, Príncipe de Asturias, University Hospital, 28805 Alcala de Henares, Spain
| | - Cielo Garcia-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Pathological Anatomy Service, Central University Hospital of Defence—UAH Madrid, 28801 Alcala de Henares, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, 28806 Alcala de Henares, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Department of General and Digestive Surgery, Príncipe de Asturias, University Hospital, 28805 Alcala de Henares, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| |
Collapse
|
39
|
Liu C, Yin Q, Wu Z, Li W, Huang J, Chen B, Yang Y, Zheng X, Zeng L, Wang J. Inflammation and Immune Escape in Ovarian Cancer: Pathways and Therapeutic Opportunities. J Inflamm Res 2025; 18:895-909. [PMID: 39867950 PMCID: PMC11762012 DOI: 10.2147/jir.s503479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/06/2025] [Indexed: 01/28/2025] Open
Abstract
Ovarian cancer (OC) remains one of the most lethal gynecological malignancies, largely due to its late-stage diagnosis and high recurrence rates. Chronic inflammation is a critical driver of OC progression, contributing to immune evasion, tumor growth, and metastasis. Inflammatory cytokines, including IL-6, TNF-α, and IL-8, as well as key signaling pathways such as nuclear factor kappa B (NF-kB) and signal transducer and activator of transcription 3 (STAT3), are upregulated in OC, promoting a tumor-promoting environment. The tumor microenvironment (TME) is characterized by immune cells like tumor-associated macrophages (TAMs) and regulatory T cells (Tregs), which suppress anti-tumor immune responses, facilitating immune evasion. Furthermore, OC cells utilize immune checkpoint pathways, including PD-1/PD-L1, to inhibit cytotoxic T cell activity. Targeting these inflammatory and immune evasion mechanisms offers promising therapeutic strategies. COX-2 inhibitors, Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway blockers, and NF-kB inhibitors have shown potential in preclinical studies, while immune checkpoint inhibitors targeting PD-1/PD-L1 and CTLA-4 have been explored with mixed results in OC. Additionally, emerging research on the microbiome and inflammation-related biomarkers, such as microRNAs (miRNAs) and exosomes, points to new opportunities for early detection and precision medicine. Future approaches to OC treatment must focus on personalized strategies that target the inflammatory TME, integrating anti-inflammatory therapies with immunotherapy to enhance patient outcomes. Continued research into the interplay between inflammation and immune evasion in OC is essential for developing effective, long-lasting treatments.
Collapse
Affiliation(s)
- Chunyan Liu
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Qinan Yin
- Department of Radiation Oncology, First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, People’s Republic of China
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Zhaoying Wu
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Wenhui Li
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Jun Huang
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Bo Chen
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Yanjun Yang
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Xuewei Zheng
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Li Zeng
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Jingjing Wang
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, People’s Republic of China
| |
Collapse
|
40
|
Sawaya M, Cordina-Duverger E, Lamy PJ, Trétarre B, Menegaux F. Kidney and gallbladder stones and the risk of prostate cancer: Results from the EPICAP study. PLoS One 2025; 20:e0317760. [PMID: 39823409 PMCID: PMC11741565 DOI: 10.1371/journal.pone.0317760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/04/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Prostate cancer remains the most frequent cancer among men, representing a significant health burden. Despite its high morbidity and mortality rates, the etiology of prostate cancer remains relatively unknown, with only non-modifiable established risk factors. Chronic inflammation has emerged as a potential factor in prostate carcinogenesis. We investigated the role of kidney and gallbladder stones and the risk of prostate cancer. METHODS We used data from EPICAP, a population-based case-control study. A total of 819 diagnosed prostate cancer cases and 879 controls were face-to-face interviewed using a standardized questionnaire that collected information on personal medical history, including history of kidney and gallbladder stones. Odds Ratios (OR) and their 95% confidence interval (CI) were estimated using multivariate unconditional logistic regression. RESULTS Our study revealed intriguing patterns regarding kidney and gallbladder stones in relation to prostate cancer risk. The analysis indicated significant potential associations between kidney stones and the risk of prostate cancer (OR: 1.46 95% CI: 1.13-1.90), particularly in men with a history of kidney infection. Additionally, our data suggested a possible relationship between gallbladder stones and prostate cancer when considering triglyceride (OR: 2.27, 95% CI: 0.99-5.28), although further research is needed for a conclusive understanding. CONCLUSIONS Our results suggest an association between calculi and the risk of prostate cancer. Findings from this study underscore the need for a more comprehensive investigation to understand the role of chronic inflammation or metabolism and delineate the mechanisms underlying these potential associations in order to guide the development of targeted preventive strategies for aggressive prostate cancer.
Collapse
Affiliation(s)
- Melissa Sawaya
- UVSQ, Inserm, Gustave Roussy, CESP, Université Paris-Saclay, Villejuif, France
| | | | - Pierre-Jean Lamy
- Service de Recherche Clinique, Clinique Beau Soleil, Montpellier, France
- Service Urologie, Clinique Beau Soleil, Montpellier, France
| | - Brigitte Trétarre
- Registre des Tumeurs de l’Hérault, EA 2415, ICM, Montpellier, France
- Center for Epidemiology and Research in Population Health (CERPOP), Toulouse, France
| | - Florence Menegaux
- UVSQ, Inserm, Gustave Roussy, CESP, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
41
|
Fang S, Jiang M, Jiao J, Zhao H, Liu D, Gao D, Wang T, Yang Z, Yuan H. Unraveling the ROS-Inflammation-Immune Balance: A New Perspective on Aging and Disease. Aging Dis 2025:AD.2024.1253. [PMID: 39812539 DOI: 10.14336/ad.2024.1253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025] Open
Abstract
Increased entropy is a common cause of disease and aging. Lifespan entropy is the overall increase in disorder caused by a person over their lifetime. Aging leads to the excessive production of reactive oxygen species (ROS), which damage the antioxidant system and disrupt redox balance. Organ aging causes chronic inflammation, disrupting the balance of proinflammatory and anti-inflammatory factors. Inflammaging, which is a chronic low-grade inflammatory state, is activated by oxidative stress and can lead to immune system senescence. During this process, entropy increases significantly as the body transitions from a state of low order to high disorder. However, the connection among inflammation, aging, and immune system activity is still not fully understood. This review introduces the idea of the ROS-inflammation-immune balance for the first time and suggests that this balance may be connected to aging and the development of age-related diseases. We also explored how the balance of these three factors controls and affects age-related diseases. Moreover, imbalance in the relationship described above disrupts the regular structures of cells and alters their functions, leading to cellular damage and the emergence of a disorganized state marked by increased entropy. Maintaining a low entropy state is crucial for preventing and reversing aging processes. Consequently, we examined the current preclinical evidence for antiaging medications that target this balance. Ultimately, comprehending the intricate relationships between these three factors and the risk of age-related diseases in organisms will aid in the development of clinical interventions that promote long-term health.
Collapse
Affiliation(s)
- Sihang Fang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Mingjun Jiang
- Respiratory Department, Beijing Children's Hospital, Capital Medical University, China National Clinical Research Center of Respiratory Diseases, National Center for Children's Health, Beijing, China
| | - Juan Jiao
- Department of Clinical Laboratory, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Hongye Zhao
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dizhi Liu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Danni Gao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Tenger Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Ze Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Huiping Yuan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| |
Collapse
|
42
|
Lee J, Woo H, Kang H, Park YK, Lee JY. Nicotinamide riboside targets mitochondrial unfolded protein response to reduce alcohol-induced damage in Kupffer cells. J Pathol 2025; 265:110-122. [PMID: 39624887 DOI: 10.1002/path.6372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 09/15/2024] [Accepted: 10/23/2024] [Indexed: 12/14/2024]
Abstract
The pathogenesis of alcohol-related liver disease (ALD) is closely linked to mitochondrial dysfunction and impaired cellular energy metabolism. In this study, we explored how ethanol triggers inflammation, oxidative stress, and mitochondrial dysfunction in Kupffer cells, i.e.hepatic resident macrophages, primarily focusing on the mitochondrial unfolded protein response (UPRmt) using immortalized mouse Kupffer cells (ImKCs) and mouse primary KCs. The UPRmt is a cellular defense mechanism activated in response to the perturbation of mitochondrial proteostasis to maintain mitochondrial integrity and function by upregulating the expression of mitochondrial chaperones and proteases. We also determined whether nicotinamide riboside (NR), a NAD+ precursor, could mitigate ethanol-triggered cellular damage. When ImKCs were exposed to 80 mm ethanol for 72 h, they displayed inflammation, oxidative stress, and impaired mitochondrial function with decreased mitochondrial content and deformed mitochondrial crista structure. NR, however, counteracted the effects of ethanol. Furthermore, ethanol increased mRNA and protein levels of UPRmt genes, such as mitochondrial chaperones and proteases, which were attenuated by NR. Notably, the ethanol-induced shift in the entry of activating transcription factor 5 (ATF5), a putative transcriptional regulator of UPRmt, to the nucleus from the mitochondria was abolished by NR. The induction of UPRmt genes by ethanol was significantly repressed when Atf5 was knocked down, indicating the role of ATF5 in the induction of UPRmt genes in ImKCs exposed to ethanol. We also confirmed the induction of UPRmt gene expression in mouse and human livers exposed to alcohol. Our findings demonstrate the ability of NR to alleviate ethanol-induced oxidative stress, inflammation, and mitochondrial dysfunction, partly by modulating the ATF5-dependent UPRmt pathway in ImKCs, suggesting its potential for ALD therapy. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jaeeun Lee
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, USA
| | - Hayoung Woo
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, USA
| | - Hyunju Kang
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, USA
- Department of Food and Nutrition, Keimyung University, Daegu, South Korea
| | - Young-Ki Park
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, USA
| | - Ji-Young Lee
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
43
|
Sakaguchi R, Sasaki K, Oshima J, Aihara Y, Sekido M. Pedicled Latissimus Dorsi Flap Vascularized by a Lumbar Artery Perforator for the Reconstruction of an Exposed Lumbar Spinal Fixation Device: A Case Report. Cureus 2025; 17:e78163. [PMID: 40026939 PMCID: PMC11870773 DOI: 10.7759/cureus.78163] [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] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Latissimus dorsi flaps are typically vascularized by the thoracodorsal or intercostal artery perforators, while cases describing the use of lumbar artery perforators are exceedingly rare. This report presents a case of a 74-year-old woman presenting with a refractory ulcer associated with an exposed lumbar spinal fixation device. Reconstruction was successfully performed using a pedicled latissimus dorsi flap vascularized by lumbar artery perforators. Preoperative indocyanine green (ICG) fluorescence angiography confirmed adequate perfusion from the lumbar artery perforators. Postoperative outcomes were favorable, with no ulcer recurrence observed after six months of follow-up. Chronic inflammation and malignancy were considered as likely contributors to angiogenesis and increased blood flow around the lumbar artery perforators. Additionally, ICG fluorescence angiography proved an effective and minimally invasive technique for evaluating flap perfusion.
Collapse
Affiliation(s)
- Riko Sakaguchi
- Department of Plastic, Reconstructive, and Hand Surgery, University of Tsukuba, Tsukuba, JPN
| | - Kaoru Sasaki
- Department of Plastic, Reconstructive, and Hand Surgery, University of Tsukuba, Tsukuba, JPN
| | - Junya Oshima
- Department of Plastic, Reconstructive, and Hand Surgery, University of Tsukuba, Tsukuba, JPN
| | - Yukiko Aihara
- Department of Plastic, Reconstructive, and Hand Surgery, University of Tsukuba, Tsukuba, JPN
| | - Mitsuru Sekido
- Department of Plastic, Reconstructive, and Hand Surgery, University of Tsukuba, Tsukuba, JPN
| |
Collapse
|
44
|
Malhotra K, Malik A, Almalki WH, Sahebkar A, Kesharwani P. Reactive Oxygen Species and its Manipulation Strategies in Cancer Treatment. Curr Med Chem 2025; 32:55-73. [PMID: 37303173 DOI: 10.2174/0929867330666230609110455] [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: 04/01/2023] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023]
Abstract
Cancer is one of the serious diseases of modern times, occurring in all parts of the world and shows a wide range of effects on the human body. Reactive Oxygen Species (ROS) such as oxide and superoxide ions have both advantages and disadvantages during the progression of cancer, dependent on their concentration. It is a necessary part of the normal cellular mechanisms. Changes in its normal level can cause oncogenesis and other relatable problems. Metastasis can also be controlled by ROS levels in the tumor cells, which can be prevented by the use of antioxidants. However, ROS is also used for the initiation of apoptosis in cells by different mediators. There exists a cycle between the production of oxygen reactive species, their effect on the genes, role of mitochondria and the progression of tumors. ROS levels cause DNA damage by the oxidation process, gene damage, altered expression of the genes and signalling mechanisms. They finally lead to mitochondrial disability and mutations, resulting in cancer. This review summarizes the important role and activity of ROS in developing different types of cancers like cervical, gastric, bladder, liver, colorectal and ovarian cancers.
Collapse
Affiliation(s)
- Kabil Malhotra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Arzoo Malik
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| |
Collapse
|
45
|
Jain P, Mohapatra S, Farooq U, Hassan N, Mirza MA, Iqbal Z. An Overview of the Dichotomous Role of Microbiota in Cancer Progression and Management. Curr Cancer Drug Targets 2025; 25:38-48. [PMID: 38409691 DOI: 10.2174/0115680096282503240124104029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 02/28/2024]
Abstract
It is a well-known fact that cancer is considered the second leading cause of mortality across the globe. Although the human oral cavity and intestine are the natural habitat of thousands of microbes, dysbiosis results in malignancies, such as oral squamous cell carcinoma and colorectal cancer. Amongst the intestinal microbes, H. pylori is a deadly carcinogen. Also, causative pathogens for the development of pancreatic and colorectal cancer are found in the oral cavity, such as Fusobacterium nucleatum and Porphyromonas gingivalis. Many periodontopathic micro- organisms, like Streptococcus sp., Peptostreptococcus sp., Prevotella sp., Fusobacterium sp., Porphyromonas gingivalis, and Capnocytophaga gingivalis, strongly have an impact on the development of oral cancers. Three basic mechanisms are involved in pathogen-mediated cancer development, like chronic inflammation-mediated angiogenesis, inhibition of cellular apoptosis, and release of carcinogenic by-products. Microbiota has a dichotomous role to play in cancer, i.e., microbiota can be used for cancer management too. Shreds of evidence are there to support the fact that microbiota enhances the chemotherapeutic drug efficacy. This review presents the possible mechanism of the oncogenic effect of microbiota with emphasis on the oral microbiome and also attempts to explain the intricate role of microbiota in cancer management.
Collapse
Affiliation(s)
- Pooja Jain
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110030, India
| | - Sradhanjali Mohapatra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110030, India
| | - Uzma Farooq
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110030, India
| | - Nazia Hassan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110030, India
| | - Mohd Aamir Mirza
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110030, India
| | - Zeenat Iqbal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110030, India
| |
Collapse
|
46
|
Huang Z, Wang G, Xu L, Cui S, Sun X, Li T, Wang J, Xu M, Xie M. Prognostic value of the preoperative ratio of fibrinogen to prealbumin in patients undergoing radical resection for lung cancer. ANZ J Surg 2025; 95:91-99. [PMID: 39927605 DOI: 10.1111/ans.19391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 06/21/2024] [Accepted: 01/01/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND To investigate the significance of preoperative fibrinogen-to-prealbumin ratio (FPR) in the prognosis of non-small cell lung cancer. METHODS The clinical follow-up pathological data of 289 patients who underwent radical lung cancer resection and were pathologically diagnosed with non-small cell lung cancer after surgery were retrospectively analysed, and the FPR value was calculated according to the serological test results within 1 week before surgery. The cut-off value of FPR was obtained by analysing the receiver operating characteristic curve (ROC). Kaplan-Meier analysis was used to describe the survival curve, and Cox proportional hazards model was used to explore the risk factors affecting prognosis. RESULTS When the FPR value was 10.96, the Youden index was the highest, with a sensitivity of 62.4% and a specificity of 69.6%. The cumulative five-year survival rate in the low FPR group was significantly higher than that in the high FPR group (81.4% versus 54.7%). Univariate and multivariate analyses showed that FPR level was a risk factor for prognosis. CONCLUSION There is a correlation between FPR level and the prognosis of lung cancer patients, and early intervention should be implemented for patients with high FPR before surgery.
Collapse
Affiliation(s)
- Zhining Huang
- Department of Thoracici Suegery, Anhui Medical University Affiliated Provincial Hospital, Hefei, China
| | - Gaoxiang Wang
- Department of Thoracici Suegery, Anhui Medical University Affiliated Provincial Hospital, Hefei, China
| | - Liangdong Xu
- Department of Thoracici Suegery, Anhui Medical University Affiliated Provincial Hospital, Hefei, China
| | - Shijun Cui
- Department of Thoracici Suegery, Anhui Medical University Affiliated Provincial Hospital, Hefei, China
| | - Xiaohui Sun
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Tian Li
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Jun Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Meiqing Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Mingran Xie
- Department of Thoracici Suegery, Anhui Medical University Affiliated Provincial Hospital, Hefei, China
| |
Collapse
|
47
|
Ayub MA, Tyagi AR, Srivastava SK, Singh P. Quantum DFT analysis and molecular docking investigation of various potential breast cancer drugs. J Mater Chem B 2024; 13:218-238. [PMID: 39545283 DOI: 10.1039/d4tb01803f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Breast cancer is among the deadliest cancers worldwide, highlighting the urgent need for effective treatments. This study employs density functional theory (DFT) and molecular docking analyses to evaluate the anti-cancer efficacy and specificity of drug molecules lapatinib, tucatinib, neratinib, anastrozole, and letrozole. DFT analysis provides comprehensive insights into the structural, electronic, optical, and vibrational properties of these drugs, helping to elucidate their molecular stability and reactivity through global reactivity descriptors. Additionally, molecular docking simulations reveal the binding conformations and interaction profiles of these drugs with key breast cancer targets, underscoring their therapeutic potential. Docking results indicate that lapatinib, tucatinib, and neratinib have high binding affinities for HER2, with lapatinib exhibiting the strongest overall binding, particularly with PDK1 (PDB ID: 1UU7), PAK4 (PDB ID: 2X4Z), GSK3 (PDB ID: 1GNG), and HER2 (PDB ID: 2IOK). The stable hydrogen bonding and other interactions observed with lapatinib support its effectiveness in treating HER2-positive breast cancers, tucatinib's selective HER2 binding reduces off-target effects, while neratinib's irreversible binding provides prolonged inhibition, making it useful for overcoming resistance in HER2-positive cases. In contrast, anastrozole and letrozole show lower binding affinities for HER2 and EGFR due to their simpler structures but are potent aromatase inhibitors, making them effective in treating estrogen receptor-positive (ER-positive) breast cancers. In conclusion, DFT and molecular docking studies affirm the suitability of lapatinib, tucatinib, and neratinib for HER2-positive cancers, while anastrozole and letrozole are effective in ER-positive cancers, emphasizing the role of molecular structure and binding affinity in optimizing cancer treatment strategies.
Collapse
Affiliation(s)
- Md Ashraf Ayub
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari-845401, Bihar, India.
| | - Ankit Raj Tyagi
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari-845401, Bihar, India.
| | - Sunil Kumar Srivastava
- Department of Physics, School of Physical Sciences, Mahatma Gandhi Central University, Motihari-845401, Bihar, India.
| | - Pranveer Singh
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari-845401, Bihar, India.
| |
Collapse
|
48
|
Xie H, Xi X, Lei T, Liu H, Xia Z. CD8 + T cell exhaustion in the tumor microenvironment of breast cancer. Front Immunol 2024; 15:1507283. [PMID: 39717767 PMCID: PMC11663851 DOI: 10.3389/fimmu.2024.1507283] [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: 10/07/2024] [Accepted: 11/20/2024] [Indexed: 12/25/2024] Open
Abstract
CD8+ T cells are crucial cytotoxic components of the tumor immune system. In chronic inflammation, they become low-responsive, a state known as T cell exhaustion (TEX). The aim of immune checkpoint blockade is to counteract TEX, yet its dynamics in breast cancer remain poorly understood. This review defines CD8+ TEX and outlines its features and underlying mechanisms. It also discusses the primary mechanisms of CD8+ TEX in breast cancer, covering inhibitory receptors, immunosuppressive cells, cytokines, transcriptomic and epigenetic alterations, metabolic reprogramming, and exosome pathways, offering insights into potential immunotherapy strategies for breast cancer.
Collapse
Affiliation(s)
- Hanghang Xie
- Xi’an People’s Hospital (Xi’an Fourth Hospital), Affiliated People’s Hospital of Northwest University, Xi’an, China
| | - Xiaowei Xi
- Technical University of Munich (TUM) School of Medicine and Health, Munich, Germany
| | - Ting Lei
- Xi’an People’s Hospital (Xi’an Fourth Hospital), Affiliated People’s Hospital of Northwest University, Xi’an, China
| | - Hongli Liu
- Xi’an People’s Hospital (Xi’an Fourth Hospital), Affiliated People’s Hospital of Northwest University, Xi’an, China
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians University, Munich, Germany
| |
Collapse
|
49
|
Mercuţ R, Ciurea ME, Traşcă ET, Ionescu M, Mercuţ MF, Rădulescu PM, Călăraşu C, Streba L, Ionescu AG, Rădulescu D. Applying Neural Networks to Analyse Inflammatory, Sociodemographic, and Psychological Factors in Non-Melanoma Skin Cancer and Colon Cancer: A Statistical and Artificial Intelligence Approach. Diagnostics (Basel) 2024; 14:2759. [PMID: 39682667 DOI: 10.3390/diagnostics14232759] [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: 11/02/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: Chronic inflammation and psychosocial factors significantly influence cancer progression and patient behavior in seeking medical care. Understanding their interplay is essential for enhancing early detection and developing personalized treatment strategies. This study aims to develop a comprehensive patient profiling model by comparing non-melanoma skin cancer (NMSC) and colorectal cancer (CRC). The goal is to identify common and distinct patterns in inflammation and psychosocial factors that affect disease progression and clinical presentation. Methods: We conducted a comparative analysis of patients diagnosed with NMSC and CRC, integrating clinical data with sociodemographic and psychological assessments. Advanced neural network algorithms were employed to detect subtle patterns and interactions among these factors. Based on the analysis, a cancer risk assessment questionnaire was developed to stratify patients into low-, moderate-, and high-risk categories. Results: Patients with low systemic inflammation and adequate vagal tone, supported by a stable family environment, demonstrated heightened sensitivity to subclinical symptoms, enabling earlier diagnosis and timely intervention. Conversely, patients with high systemic inflammation and reduced vagal tone, often influenced by chronic stress and unstable family environments, presented at more advanced disease stages. The developed risk assessment tool effectively classified patients into distinct risk categories, facilitating targeted preventive measures and personalized therapeutic strategies. Neural network profiling revealed significant interactions between biological and psychosocial factors, enhancing our understanding of their combined impact on cancer progression. Conclusions: The integrated profiling approach and the newly developed risk assessment questionnaire have the potential to transform cancer management by improving early detection, personalizing treatment strategies, and addressing psychosocial factors. This model not only enhances clinical outcomes and patient quality of life but also offers a framework adaptable to other cancer types, promoting a holistic and patient-centered approach in oncology.
Collapse
Affiliation(s)
- Răzvan Mercuţ
- Department of Plastic and Reconstructive Surgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Marius Eugen Ciurea
- Department of Plastic and Reconstructive Surgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Emil Tiberius Traşcă
- The Surgery Clinic of "Dr. Ștefan Odobleja Emergency Military Hospital", General Surgery Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Mihaela Ionescu
- Department of Medical Informatics and Biostatistics, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Maria Filoftea Mercuţ
- Department of Ophthalmology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | | | - Cristina Călăraşu
- Department of Pneumology, University of Pharmacy and Medicine Craiova, 200349 Craiova, Romania
| | - Liliana Streba
- Department of Oncology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Alin Gabriel Ionescu
- Department of Medical History, University of Pharmacy and Medicine Craiova, 200349 Craiova, Romania
| | - Dumitru Rădulescu
- The Surgery Clinic of "Dr. Ștefan Odobleja Emergency Military Hospital", General Surgery Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
50
|
Han C, Zhang R, He X, Fang Y, Cen G, Wu W, Huang C, Chen X. A digital manufactured microfluidic platform for flexible construction of 3D co-culture tumor model with spatiotemporal resolution. Biofabrication 2024; 17:015029. [PMID: 39577087 DOI: 10.1088/1758-5090/ad9636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/22/2024] [Indexed: 11/24/2024]
Abstract
The specific spatiotemporal distribution of diverse components in tumor microenvironment plays a crucial role in the cancer progression.In vitrothree-dimensional (3D) tumor models with polydimethylsiloxane (PDMS) based microfluidic platform have been applied as useful tool to conduct studies from cancer biology to drug screening. However, PDMS has not been welcomed as a standardized commercial application for preclinical screening due to inherent limitations in scale-up production and molecule absorption. Here, we present a novel microfluidic platform to flexibly construct 3D co-culture models with spatiotemporal resolution by using multiple digital manufacturing technologies. The platform, which consist of reduplicative microfluidic chips, is made of biocompatible poly methyl methacrylate by fast laser cutting. Each replica includes a simple microfluidic chamber without internal structures which can be flexibly post-fabricated according to various research requirements. Digital light processing based 3D bioprinting was used to pattern fine hydrogel structures for post-fabrication on-chip. By multi-step bioprinting and automatic image alignment, we show that this approach provides sufficient design flexibility to construct 3D co-culture tumor model with spatiotemporal resolution to replicate microarchitecture of tumor microtissuein situ. And the tumor model has the potential to mimic tumor biology behaviors which can be used for mechanism study and drug test. Our microengineered tumor model may serve as an enabling tool to recapitulate pathophysiological complexity of tumor, and to systematically examine the contribution of the tumor microenvironment to the cancer progression. The proposed strategy can also be applied to help engineer diverse meaningfulin vitromodels for extensive biomedical applications, from physiology and disease study to therapy evaluation.
Collapse
Affiliation(s)
- Chao Han
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
- Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Renchao Zhang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Xiwen He
- Aerospace Laser Technology and Systems Department, Shanghai Institute of Optics and Fine Mechanics, Chinese Academy of Sciences, Shanghai 201800, People's Republic of China
| | - Yuan Fang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Gang Cen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Weidong Wu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Chen Huang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Xiang Chen
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
- Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| |
Collapse
|