1
|
Wang M, Wan Q, Wang C, Jing Q, Nie Y, Zhang X, Chen X, Yang D, Pan R, Li L, Zhu L, Gui H, Chen S, Deng Y, Chen T, Nie Y. Combinational delivery of TLR4 and TLR7/8 agonist enhanced the therapeutic efficacy of immune checkpoint inhibitors to colon tumor. Mol Cell Biochem 2025; 480:445-458. [PMID: 38507020 DOI: 10.1007/s11010-024-04966-6] [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: 12/24/2023] [Accepted: 02/11/2024] [Indexed: 03/22/2024]
Abstract
Immunotherapy is regarded as a potent cancer treatment, with DC vaccines playing a crucial role. Although clinical trials have demonstrated the safety and efficacy of DC vaccines, loading antigens in vitro is challenging, and their therapeutic effects remain unpredictable. Moreover, the diverse subtypes and maturity states of DCs in the body could induce both immune responses and immune tolerance, potentially affecting the vaccine's efficacy. Hence, the optimization of DC vaccines remains imperative. Our study discovered a new therapeutic strategy by using CT26 and MC38 mouse colon cancer models, as well as LLC mouse lung cancer models. The strategy involved the synergistic activation of DCs through intertumoral administration of TLR4 agonist high-mobility group nucleosome binding protein 1 (HMGN1) and TLR7/8 agonist (R848/resiquimod), combined with intraperitoneal administration of TNFR2 immunosuppressant antibody. The experimental results indicated that the combined use of HMGN1, R848, and α-TNFR2 had no effect on LLC cold tumors. However, it was effective in eradicating CT26 and MC38 colon cancer and inducing long-term immune memory. The combination of these three drugs altered the TME and promoted an increase in anti-tumor immune components. This may provide a promising new treatment strategy for colon cancer.
Collapse
Affiliation(s)
- Mengjiao Wang
- GuiZhou University Medical College, Guiyang, 550025, China
| | - Quan Wan
- School of Preclinical Medicine of Zunyi Medical University, Zunyi, 563000, China
| | - Chenglv Wang
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Qianyu Jing
- School of Preclinical Medicine of Zunyi Medical University, Zunyi, 563000, China
| | - Yujie Nie
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Xiangyan Zhang
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, SAR, China
| | - De Yang
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute at Frederick, NIH, Frederick, MD, USA
| | - Runsang Pan
- Department of Pathophysiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - Linzhao Li
- GuiZhou University Medical College, Guiyang, 550025, China
| | - Lan Zhu
- GuiZhou University Medical College, Guiyang, 550025, China
| | - Huan Gui
- GuiZhou University Medical College, Guiyang, 550025, China
| | - Shuanghui Chen
- GuiZhou University Medical College, Guiyang, 550025, China
| | - Yuezhen Deng
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Tao Chen
- State Key Laboratory of Respiratory Disease at People's Hospital of Yangjiang, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| | - Yingjie Nie
- Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China.
| |
Collapse
|
2
|
Mariana SM, Brenda RP, Heriberto PG, Cristina L, David B, Guadalupe ÁL. GPER1 activation by estrogenic compounds in the inflammatory profile of breast cancer cells. J Steroid Biochem Mol Biol 2025; 245:106639. [PMID: 39571822 DOI: 10.1016/j.jsbmb.2024.106639] [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: 08/08/2024] [Revised: 10/28/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Breast cancer (BC) is the most frequent female neoplasm worldwide. Its establishment and development have been related to inflammatory cytokine expression. Steroid hormones such as estradiol (E2) can regulate proinflammatory cytokine secretion through interaction with its nuclear receptors. However, little is known regarding the activation of its membrane estrogen receptor (GPER1) and the inflammatory cytokine environment in BC. We have studied the synthesis and biological effects of molecules analogs to E2 for hormone replacement therapy (HRT), such as pentolame. Nevertheless, its interaction with GPER1 and the modulation of inflammatory cytokines in different BC types has been barely studied and deserves deeper investigation. In this research, the role of GPER1 in the proliferation and modulation of inflammatory cytokines involved in carcinogenesis and metastatic processes in different BC cell lines was assessed by binding to various compounds. To achieve this goal, the presence of GPER1 was identified in different BC cell lines. Subsequently, cell proliferation after exposure to E2, pentolame and GPER1 agonist, G1, was subsequently determined alone or in combination with the GPER1 antagonist, G15. Finally, the pro-inflammatory cytokine secretion derived from the supernatants of BC cells exposed to the previous treatments was also assessed. Interestingly, GPER1 activation or inhibition has significant effects on the cytokine regulation associated with invasion in BC. Notably, pentolame did not induce cell proliferation or increase the proinflammatory cytokine expression compared to E2 in BC cell lines. In addition, pentolame did not induce the presence of the cell adhesion molecule PECAM-1. In contrast, E2 treatment weakly induced the expression of PECAM-1 in MCF-7 and HCC1937 cells, and G1 treatment showed this effect only in MCF-7 cells. The results suggest that GPER1 might be a significant inflammatory modulator with angiogenic-related effects in BC cells. In addition, pentolame might represent an HRT alternative in patients with BC predisposition.
Collapse
Affiliation(s)
- Segovia-Mendoza Mariana
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| | - Reyes-Plata Brenda
- Facultad de Estudios Superiores Zaragoza. Universidad Nacional Autónoma de México,Ciudad de México, Mexico
| | - Prado-Garcia Heriberto
- Laboratorio de Onco-Inmunobiología, Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias, "Ismael Cosio Villegas" Calzada de Tlalpan 4502, Col. Sección XVI, Ciudad de México 14080, Mexico
| | - Lemini Cristina
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Barrera David
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Col. Belisario Domínguez, Sección XVI, Ciudad de México 14080, Mexico
| | - Ángeles-López Guadalupe
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| |
Collapse
|
3
|
Kravtsova-Ivantsiv Y, Goldhirsh G, Ciechanover A. CXCL12 restricts tumor growth by suppressing the Ras, ERK1/2, c-Myc, and the immune checkpoint PD-L1 pathways. Proc Natl Acad Sci U S A 2024; 121:e2416909121. [PMID: 39689179 DOI: 10.1073/pnas.2416909121] [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: 08/20/2024] [Accepted: 11/15/2024] [Indexed: 12/19/2024] Open
Abstract
Cytokines constitute a family of proteins that modulate the immune system and are secreted by many cells. CXCL12, along with its receptor CXCR4, are essential players in numerous processes. Dysregulation of their function underlie the mechanism(s) of several pathologies, including malignancies. Here, we demonstrate an unexpected effect of the cytokine and its receptor: In both cells and animal models, CXCL12 restricts tumorigenicity of the human glioblastoma cells U87-MG and U-118, and of a cell line derived from PyMT mouse breast cancer. Overexpression of CXCL12 inhibits activation of the oncogene Ras which results in downregulation of its proliferative signals, such as reduced phosphorylation of the extracellular signal-regulated kinase 1/2 (ERK1/2), inhibition of c-Myc expression, and subsequent inhibition of cell cycle. Furthermore, CXCL12 induces downregulation of the growth differentiation factor 15 (GDF15), insulin-like growth factor-binding protein 6 (IGFBP6), and matrix metalloproteinase-3 (MMP3), which are implicated in sending metastases. Indeed, monitoring cell migration in vitro and generation of metastases in mice demonstrate that CXCL12 slows the migration of U87-MG and PyMT cells. Remarkably, overexpression of CXCL12 also downregulates the cell surface immune checkpoint protein programmed cell death-ligand 1 (PD-L1), resulting in recruitment of cytotoxic CD8 T cells into xenografts accompanied by their shrinkage. Overall, CXCL12 inhibits tumor growth through several distinct mechanisms: inhibition of cell cycle and migration, as well as impairment of immune checkpoint, thereby stimulating a strong host's immune response. The mechanism(s) that renders CXCL12 a tumor-promoting factor in certain cells and a suppressor in others has remained elusive.
Collapse
Affiliation(s)
- Yelena Kravtsova-Ivantsiv
- The Rappaport Faculty of Medicine and Research Institute, and the Rappaport Technion Integrated Cancer Center (R-TICC), Technion-Israel Institute of Technology, Haifa 3109601, Israel
| | - Gilad Goldhirsh
- The Rappaport Faculty of Medicine and Research Institute, and the Rappaport Technion Integrated Cancer Center (R-TICC), Technion-Israel Institute of Technology, Haifa 3109601, Israel
| | - Aaron Ciechanover
- The Rappaport Faculty of Medicine and Research Institute, and the Rappaport Technion Integrated Cancer Center (R-TICC), Technion-Israel Institute of Technology, Haifa 3109601, Israel
| |
Collapse
|
4
|
Guo X, Zuo Z, Wang X, Sun Y, Xu D, Liu G, Tong Y, Zhang Z. Epidemiology, risk factors and mechanism of breast cancer and atrial fibrillation. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:92. [PMID: 39716319 DOI: 10.1186/s40959-024-00298-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
Cancer and cardiovascular diseases are leading causes of death worldwide. Among them, breast cancer is one of the most common malignancies in women, while atrial fibrillation is one of the most extensively studied arrhythmias, with significant public health implications. As the global population ages and advancements in cancer treatments continue, the survival rates of breast cancer patients have significantly improved, leading to an increasing coexistence of breast cancer and atrial fibrillation. However, the mechanisms underlying this coexistence remain insufficiently studied, and there is no consensus on the optimal treatment strategies for these patients. This review consolidates existing research to systematically explore the epidemiological characteristics, risk factors, and pathophysiological mechanisms of both breast cancer and atrial fibrillation. It focuses on the unique signaling pathways associated with different molecular subtypes of breast cancer and their potential impact on the mechanisms of atrial fibrillation. Additionally, the relationship between atrial fibrillation treatment medications and breast cancer is discussed. These insights not only provide essential evidence for the precise prevention and management of atrial fibrillation in breast cancer patients but also lay a solid theoretical foundation for interdisciplinary clinical management practices.
Collapse
Affiliation(s)
- Xiaoxue Guo
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Zheng Zuo
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Xishu Wang
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Ying Sun
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Dongyang Xu
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Guanghui Liu
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yi Tong
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Zhiguo Zhang
- Department of Cardiology, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin Province, China.
| |
Collapse
|
5
|
Wang Y, Zheng Y, Tian C, Yu J, Rao K, Zeng N, Jiang P. Nomogram Based on Immune-Inflammatory Score and Classical Clinicopathological Parameters for Predicting the Recurrence of Endometrial Carcinoma: A Large, Multi-Center Retrospective Study. J Inflamm Res 2024; 17:11437-11449. [PMID: 39735898 PMCID: PMC11675361 DOI: 10.2147/jir.s494716] [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: 09/05/2024] [Accepted: 11/28/2024] [Indexed: 12/31/2024] Open
Abstract
Background Surgery is the best approach to treat endometrial cancer (EC); however, there is currently a deficiency in effective scoring systems for predicting EC recurrence post-surgical resection. This study aims to develop a clinicopathological-inflammatory parameters-based nomogram to accurately predict the postoperative recurrence-free survival (RFS) rate of EC patients. Methods A training set containing 1068 patients and an independent validation set consisting of 537 patients were employed in this retrospective study. The prognostic factors for RFS were identified by univariable and multivariable Cox proportional hazards regression analyses, and integrated into nomogram. The C-index, area under the curves (AUC), and calibration curves were employed to determine the predictive discriminability and accuracy of nomogram. Utilizing the nomogram, patients were stratified into low- and high-risk groups, and the Kaplan-Meier survival curve was further employed to assess the clinical efficacy of the model. Results Cox regression analyses revealed that age (HR = 1.769, P = 0.002), FIGO staging (HR = 1.790, P = 0.018), LVSI (HR = 1.654, P = 0.017), Ca125 (HR = 1.532, P = 0.023), myometrial invasion (HR = 1.865, P = 0.001), cervical stromal invasion (HR = 1.655, P = 0.033), histology (HR = 2.637, P < 0.001), p53 expression (HR = 1.706, P = 0.002), PLR (HR = 1.971, P = 0.003), SIRI (HR = 2.187, P = 0.003), and adjuvant treatment (HR = 0.521, P = 0.003) were independent prognostic factors for RFS in patients with EC. A combined clinicopathologic-inflammatory parameters model was constructed, which outperformed the single-indicator model and other established models in predicting the 1-, 3-, and 5-year RFS rates in patients with EC. Conclusion The nomogram demonstrated sufficient accuracy in predicting the RFS probabilities of EC, enabling personalized clinical decision-making for future clinical endeavors.
Collapse
Affiliation(s)
- Yuqi Wang
- Department of Gynecology, Yubei District People’s Hospital, Chongqing, 401120, People’s Republic of China
| | - Yunfeng Zheng
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Chenfan Tian
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Jiaxin Yu
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Kunying Rao
- Department of Gynecology, Yubei District People’s Hospital, Chongqing, 401120, People’s Republic of China
| | - Na Zeng
- Department of Gynecology, Yubei District People’s Hospital, Chongqing, 401120, People’s Republic of China
| | - Peng Jiang
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| |
Collapse
|
6
|
Zhang Y, Tang X, Wang Y, Shi F, Gao X, Guo Y, Liu Q, Ma W. Recent advances targeting chemokines for breast cancer. Int Immunopharmacol 2024; 146:113865. [PMID: 39718056 DOI: 10.1016/j.intimp.2024.113865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/02/2024] [Accepted: 12/11/2024] [Indexed: 12/25/2024]
Abstract
Breast cancer (BC) is a complex and heterogeneous disease, and its onset and progression involve the interplay of multiple molecular mechanisms. Chemokines and their receptors are key regulators of cell migration and immune responses and contribute significantly to the pathophysiology of BC. This article reviews the classification, functions, and mechanisms of chemokines and their receptors in the proliferation, migration, invasion, and angiogenesis of BC cells. This study explores the regulatory roles of chemokines and their receptors in the immune microenvironment of BC, particularly the ways they influence the infiltration, polarization, and antitumor immune responses of immune cells. Finally, this article summarizes the current treatment strategies for breast cancer that utilize chemokines and their receptors and provides insights into future research directions and trends in this field.
Collapse
Affiliation(s)
- Yanan Zhang
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Xiufeng Tang
- Department of Pharmacy and Shandong Provincinal Key Traditional Chinese Medical Discipline of Clinical Chinese Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, China.
| | - Ying Wang
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Fengcui Shi
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Xing Gao
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Yingxin Guo
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Qian Liu
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Wenjian Ma
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
| |
Collapse
|
7
|
Liang X, Zhou J, Wang M, Wang J, Song H, Xu Y, Li Y. Progress and prospect of polysaccharides as adjuvants in vaccine development. Virulence 2024; 15:2435373. [PMID: 39601191 PMCID: PMC11622597 DOI: 10.1080/21505594.2024.2435373] [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: 09/03/2024] [Revised: 11/07/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024] Open
Abstract
Vaccines are an effective approach to confer immunity against infectious diseases. Modern subunit vaccines offer more precise target and safe protection compared to traditional whole-pathogen vaccines. However, subunit vaccines require adjuvants to stimulate the immune system due to the less immunogenicity. Adjuvants strengthen immunogenicity by enhancing, modulating, and prolonging the immune response. Unfortunately, few adjuvants have sufficient potency and low enough toxicity for clinical use, highlighting the urgent need for new vaccine adjuvants with the characteristics of safety, efficacy, and cost-effectiveness. Notably, some natural polysaccharides have been approved as adjuvants in human vaccines, owing to their intrinsic immunomodulation, low toxicity, and high safety. Natural polysaccharides are mainly derived from plants, bacteria, and yeast. Partly owing to the difficulty of obtaining them, synthetic polysaccharides emerged in clinical trials. The immune mechanisms of both natural and synthetic polysaccharides remain incompletely understood, hindering the rational development of polysaccharide adjuvants. This comprehensive review primarily focused on several promising polysaccharide adjuvants, discussing their recent applications in vaccines and highlighting their immune-modulatory effects. Furthermore, the future perspectives of polysaccharides offer insightful guidance to adjuvant development and application.
Collapse
Affiliation(s)
- Xinlong Liang
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, China
| | - Jiaying Zhou
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, China
| | - Mengmeng Wang
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, China
| | - Jing Wang
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, China
| | - Houhui Song
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, China
| | - Yigang Xu
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, China
| | - Yuan Li
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, China
- Research and Development Department, Zhejiang Huijia Biotechnology Co. Ltd ., Huzhou, People’s Republic of China
| |
Collapse
|
8
|
Zhu M, Xing M, Sun R, Li M, Qian W, Fan M. Identification of potential immune-related genes and infiltrations in temporomandibular joint osteoarthritis. Ann Med Surg (Lond) 2024; 86:7135-7146. [PMID: 39649914 PMCID: PMC11623849 DOI: 10.1097/ms9.0000000000002682] [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: 07/17/2024] [Accepted: 10/12/2024] [Indexed: 12/11/2024] Open
Abstract
Objective The aim of this study was to investigate the potential inflammatory cytokines and chemokines markers for temporomandibular joint osteoarthritis (TMJOA) diagnosis using a bioinformatics analysis. Methods The differentially expressed genes of mRNA (DEGs) and transcripts of lncRNA (DETs) were identified between TMJOA samples and normal controls curated from GSE205389 by the "DESeq. 2" R package. KEGG and GO were conducted using the R package "ggplot2" and "clusterProfiler". A PPI network was constructed to identify hub genes by using the STRING and Cytoscape. The co-expression network was constructed between mRNA and lncRNA to check the potential regulation and function of lncRNA on protein-coding genes. Finally, the immune cell infiltration analysis was conducted with CIBERSORTx and confirmed with xCells. Results The authors identified 171 DEGs and DETs, of which the DEGs were closely related to immune response, T-cell activation, cytokine-cytokine-receptor interaction, and the muscle system process. PPI network of the DEGs screened the top 10 hub genes, including IL6, IL1B, IL10, CCL2, CCL5, CXCL1, CXCL10, ICAM1, CSF1 and MMP1. Additionally, the immune cell infiltration analysis showed that CD8+ T cells, M1 macrophage and B cells infiltration were increased in TMJOA samples. Finally, the authors demonstrated that the co-expression between mRNA and lncRNA was mainly enriched in inflammatory and muscle-related pathways. Conclusions The authors found that immune and muscle system-related pathways as well as the immune infiltration played a significant role in the TMJOA development. Additionally, inflammatory cytokines and chemokines could be crucial markers for early-stage TMJOA diagnosis and personalized treatment strategies.
Collapse
Affiliation(s)
- Mengjiao Zhu
- Department of Orthodontics, Shanghai Xuhui District Dental Center, Shanghai, China
| | - Min Xing
- Dental Laboratory, Shanghai Xuhui District Dental Center, Shanghai, China
| | - Ruinan Sun
- Department of Endodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji Research Institute of Stomatology, Shanghai, China
| | - Minhui Li
- Department of vascular surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenhao Qian
- Department of Oral Implantology, Shanghai Xuhui District Dental Center, Shanghai, China
| | - Mingyue Fan
- Department of Orthodontics, Shanghai Xuhui District Dental Center, Shanghai, China
| |
Collapse
|
9
|
Hasan R, Bhuia MS, Chowdhury R, Saha S, Khan MA, Afroz M, Ansari SA, Ansari IA, Melo Coutinho HD, Islam MT. Abietic acid antagonizes the anti-inflammatory effects of celecoxib and ketoprofen: Preclinical assessment and molecular dynamic simulations. Comput Biol Med 2024; 183:109298. [PMID: 39454522 DOI: 10.1016/j.compbiomed.2024.109298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
The present work is designed to explore the anti-inflammatory properties of AA and its modulatory effects on celecoxib (CEL) and ketoprofen (KET) through in vitro, ex vivo, in vivo, and in silico approaches. Different concentrations of AA were utilized to evaluate the membrane-stabilizing potential via egg albumin and the Human Red Blood Cell (HRBC) denaturation model. In the animal model, formalin (50 μL) was injected into the right hind paw of young chicks to induce inflammation. AA was administered at 20 and 40 mg/kg (p.o.) to the experimental animals. We used CEL and KET as positive controls. The vehicle was provided as a control group. Two combinations of AA with CEL and KET were also investigated in all tests to assess the modulatory activity of AA. In addition, in silico investigation was used for predictions about drug-likeness, pharmacokinetics, and toxicity of the selected chemical compounds, and the study also evaluated the binding affinity, visualization, and stability of ligand-receptor interactions through molecular dynamic (MD) simulation. Results manifested that AA concentration-dependently significantly inhibited the egg albumin denaturation (IC50: 27.53 ± 0.88 μg/ml) and breakdown of HRBC (IC50: 15.69 ± 0.75 μg/ml), indicating the membrane stabilizing potential compared to the control group. AA also significantly (p < 0.05) lessened the frequency of licking and alleviated the paw edema in a dose-dependent manner in an in vivo test. However, AA reduced the activity of CEL and KET in combination treatment. AA showed good pharmacokinetic characteristics to be considered as a therapeutic candidate. Additionally, the in silico study displayed that AA demonstrated a relatively higher docking score of -9.1 kcal/mol with the cyclooxygenase-2 (COX-2) enzyme and stable binding in MD simulation. Whereas the standard ligand (CEL) expressed the highest binding value of -9.2 kcal/mol to the COX-2.
Collapse
Affiliation(s)
- Rubel Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh; BioLuster Research Center Ltd., Gopalgaj (Dhaka), 8100, Bangladesh.
| | - Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh; BioLuster Research Center Ltd., Gopalgaj (Dhaka), 8100, Bangladesh.
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh; BioLuster Research Center Ltd., Gopalgaj (Dhaka), 8100, Bangladesh.
| | - Sajib Saha
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh; BioLuster Research Center Ltd., Gopalgaj (Dhaka), 8100, Bangladesh.
| | - Muhammad Ali Khan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| | - Meher Afroz
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh; BioLuster Research Center Ltd., Gopalgaj (Dhaka), 8100, Bangladesh.
| | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia.
| | - Irfan Aamer Ansari
- Department of Drug Science and Technology, University of Turin, Turin, 10124, Italy.
| | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh; BioLuster Research Center Ltd., Gopalgaj (Dhaka), 8100, Bangladesh.
| |
Collapse
|
10
|
Xu J, Xu X, Zhang H, Wu J, Pan R, Zhang B. Tumor-associated inflammation: The role and research progress in tumor therapy. J Drug Deliv Sci Technol 2024; 102:106376. [DOI: 10.1016/j.jddst.2024.106376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
11
|
Liu S, Zhang X, Wang W, Li X, Sun X, Zhao Y, Wang Q, Li Y, Hu F, Ren H. Metabolic reprogramming and therapeutic resistance in primary and metastatic breast cancer. Mol Cancer 2024; 23:261. [PMID: 39574178 PMCID: PMC11580516 DOI: 10.1186/s12943-024-02165-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/31/2024] [Indexed: 11/25/2024] Open
Abstract
Metabolic alterations, a hallmark of cancer, enable tumor cells to adapt to their environment by modulating glucose, lipid, and amino acid metabolism, which fuels rapid growth and contributes to treatment resistance. In primary breast cancer, metabolic shifts such as the Warburg effect and enhanced lipid synthesis are closely linked to chemotherapy failure. Similarly, metastatic lesions often display distinct metabolic profiles that not only sustain tumor growth but also confer resistance to targeted therapies and immunotherapies. The review emphasizes two major aspects: the mechanisms driving metabolic resistance in both primary and metastatic breast cancer, and how the unique metabolic environments in metastatic sites further complicate treatment. By targeting distinct metabolic vulnerabilities at both the primary and metastatic stages, new strategies could improve the efficacy of existing therapies and provide better outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Shan Liu
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xingda Zhang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wenzheng Wang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xue Li
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xue Sun
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuqian Zhao
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qi Wang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yingpu Li
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Fangjie Hu
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| | - He Ren
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
12
|
Shen S, Qiu X, Yang C, Li J, Peng Y, Wen Z, Luo H, Xiang B. Prognostic importance of the Scottish inflammatory prognostic score in patients with hepatocellular carcinoma after hepatectomy: a retrospective cohort study. BMC Cancer 2024; 24:1393. [PMID: 39533231 PMCID: PMC11559137 DOI: 10.1186/s12885-024-13174-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: 04/11/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The Scottish Inflammatory Prognostic Score (SIPS), an innovative scoring system, has emerged as a promising biomarker for predicting patient outcomes following cancer therapy. This study aimed to evaluate the value of SIPS as a prognostic indicator following hepatectomy in patients with hepatocellular carcinoma (HCC). METHODS This retrospective study included 693 HCC patients who underwent hepatectomy. Survival outcomes were compared between propensity score-matched groups. Independent prognostic factors were identified through Cox regression analysis. Additionally, both traditional Cox proportional hazards models and machine learning models based on the SIPS were developed and validated. RESULTS A total of 693 HCC patients who underwent hepatectomy were included, with 102 in the high SIPS group and 591 in the low SIPS group. Following propensity score matching (1:3 ratio), both groups achieved balance, with 82 patients in the high SIPS group and 240 patients in the low SIPS group. The low SIPS group demonstrated significantly superior recurrence-free survival (RFS) (25 months vs. 21 months; P < 0.001) and overall survival (OS) (69 months vs. 58 months; P < 0.001) compared to the high SIPS group. Multivariable analysis identified SIPS as an independent adverse factor affecting both RFS and OS. The calibration curve for overall patient survival diagnosis displayed excellent predictive accuracy. Traditional COX prognostic models and machine learning models incorporating SIPS demonstrated excellent performance both the training and validation set. CONCLUSION This study confirms the prognostic significance of SIPS in post-hepatectomy HCC patients, providing a practical tool for risk stratification and clinical decision-making. Further research and validation are needed to consolidate its role in prognostic assessment.
Collapse
Affiliation(s)
- Shuang Shen
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning, 530021, China
| | - Xin Qiu
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Chenglei Yang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Jindu Li
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Yi Peng
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Zhaochan Wen
- Oncology School, Guangxi Medical University, Nanning, 530021, China
| | - Huili Luo
- College of Basic Medicine, Guangxi Medical University, Nanning, 530199, China
| | - Bangde Xiang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning, 530021, China.
| |
Collapse
|
13
|
Elkordy AA, Hill D, Attia M, Chaw CS. Liposomes and Their Therapeutic Applications in Enhancing Psoriasis and Breast Cancer Treatments. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1760. [PMID: 39513840 PMCID: PMC11547384 DOI: 10.3390/nano14211760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Psoriasis and breast cancer are two examples of diseases where associated inflammatory pathways within the body's immune system are implicated. Psoriasis is a complex, chronic and incurable inflammatory skin disorder that is primarily recognized by thick, scaly plaques on the skin. The most noticeable pathophysiological effect of psoriasis is the abnormal proliferation of keratinocytes. Breast cancer is currently the most diagnosed cancer and the leading cause of cancer-related death among women globally. While treatments targeting the primary tumor have significantly improved, preventing metastasis with systemic treatments is less effective. Nanocarriers such as liposomes and lipid nanoparticles have emerged as promising drug delivery systems for drug targeting and specificity. Advances in technologies and drug combinations have emerged to develop more efficient lipid nanocarriers to include more than one drug in combinational therapy to enhance treatment outcomes and/or relief symptoms for better patients' quality of life. Although there are FDA-approved liposomes with anti-cancer drugs for breast cancer, there are still unmet clinical needs to reduce the side effects associated with those nanomedicines. Hence, combinational nano-therapy may eliminate some of the issues and challenges. Furthermore, there are no nanomedicines yet clinically available for psoriasis. Hence, this review will focus on liposomes encapsulated single and/or combinational therapy to augment treatment outcomes with an emphasis on the effectiveness of combinational therapy within liposomal-based nanoparticulate drug delivery systems to tackle psoriasis and breast cancer. This review will also include an overview of both diseases, challenges in delivering drug therapy and the roles of nanomedicines as well as psoriasis and breast cancer models used for testing therapeutic interventions to pave the way for effective in vivo testing prior to the clinical trials.
Collapse
Affiliation(s)
- Amal Ali Elkordy
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK; (A.A.E.); (M.A.)
| | - David Hill
- School of Nursing and Health Sciences, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK;
| | - Mohamed Attia
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK; (A.A.E.); (M.A.)
| | - Cheng Shu Chaw
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK; (A.A.E.); (M.A.)
| |
Collapse
|
14
|
He Y, Wang D, Zhang C, Huang S, Li X, Chen Y, Ma Y, Ju S, Ye H, Fan W. EGFR-targeting oxygen-saturated nanophotosensitizers for orchestrating multifaceted antitumor responses by counteracting immunosuppressive milieu. J Control Release 2024; 375:127-141. [PMID: 39233281 DOI: 10.1016/j.jconrel.2024.08.051] [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: 06/01/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
High Epidermal growth factor receptor (EGFR) in Cutaneous Squamous Cell Carcinoma (cSCC) is associated with poor prognosis and advanced metastatic stages, severely impeding the efficacy of EGFR-targeting immunotherapy. This is commonly attributed to the combinatory outcomes of hypoxic tumor microenvironment (TME) and immunosuppressive effector cells together. Herein, a novel paradigm of EGFR-targeting oxygen-saturated nanophotosensitizers, designated as CHPFN-O2, has been specifically tailored to mitigate tumor hypoxia in EGFR-positive cSCC and achieve Cetuximab (CTX)-mediated immunotherapy (CIT). The conjugated CTX in CHPFN-O2 serves to initiate immune responses by recruiting Fc receptor (FcR)-expressing immune effector cells towards tumor cells, thereby eliciting antibody-dependent cellular phagocytosis (ADCP), antibody-dependent cellular trogocytosis (ADCT) and antibody-dependent cellular cytotoxicity (ADCC). Besides, CHPFN-O2 can engender a shift from a tumor-friendly to a tumor-hostile one through improved tumor oxygenation, contributing to oxygen-elevated photodynamic therapy (oxPDT). Notably, the combination of oxPDT and CIT eventually promotes T-cell-mediated antitumor activity and successfully inhibits the growth of EGFR-expressing cSCC with good safety profiles. This comprehensive oxPDT/CIT integration aims not only to enhance therapeutic efficacy against EGFRhigh cSCC but also to extend its applicability to other EGFRhigh malignancies, thus delineating a new avenue for the highly efficient synergistic treatment of EGFR-expressing malignancies.
Collapse
Affiliation(s)
- Yuan He
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Deng Wang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Cheng Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Siting Huang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiangzheng Li
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yue Chen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Yuanyuan Ma
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Shenghong Ju
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Hongxun Ye
- Department of Radiology, Taixing People's Hospital, Medical School, Yangzhou University, Taixing 225400, China.
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
15
|
Cullinane C, Connolly RM, Corrigan M, Redmond HP, Foley C. Perioperative systemic IL-6 and immune-adipose- metabolism transcription in tumour and tumour adjacent breast cancer. Eur J Immunol 2024; 54:e2451049. [PMID: 39219238 DOI: 10.1002/eji.202451049] [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: 02/04/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Surgical resection is the primary treatment approach for patients with breast cancer. Despite optimal multimodal treatment, metastatic recurrence remains a risk. Surgery-mediated systemic inflammation and local tissue inflammation generate an immunosuppressive and wound-healing environment that may accelerate cancer recurrence and metastasis post-operatively. Investigating the impact of surgery on local and systemic inflammation may provide knowledge for improvement of patient prognosis and treatment opportunities. Systemic cytokines were quantified in the blood plasma of patients with breast cancer pre-operatively, early post-operatively, and late post-operatively. Early post-operative levels of IL-6 were significantly elevated in patients who underwent mastectomy compared with wide local excision. Post-operative IL-6 levels correlate with clinicopathological features (age and BMI). The transcriptomes of local matched tumour and normal tumour adjacent (normal) breast tissue, from patients with breast cancer, were analysed by RNA-Seq. Elevated gene expressions of IL6, ADIPOQ, FABP4, LPL, PPARG, and CD36 in normal tissue were associated with worse overall survival of patients with ER-positive breast cancer. In tissue with higher expression of IL6 and ADIPOQ, a higher abundance of M2-like macrophage gene expression was identified. This study revealed perioperative systemic dynamics of inflammatory mediators and identified local immune-adipose-metabolism gene expression in tumour-adjacent tissue associated with pro-tumour function.
Collapse
Affiliation(s)
- Carolyn Cullinane
- Department of Surgery, School of Medicine, University College Cork, Cork, Ireland
- Cork University Hospital, Wilton, Cork, Ireland
| | - Roisin M Connolly
- Cork University Hospital, Wilton, Cork, Ireland
- Cancer Research @UCC, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Mark Corrigan
- Department of Surgery, School of Medicine, University College Cork, Cork, Ireland
- Cork University Hospital, Wilton, Cork, Ireland
- Cork Breast Research Centre, University College Cork, Cork, Ireland
| | - Henry P Redmond
- Department of Surgery, School of Medicine, University College Cork, Cork, Ireland
- Cork University Hospital, Wilton, Cork, Ireland
| | - Cathriona Foley
- Department of Surgery, School of Medicine, University College Cork, Cork, Ireland
- Cork University Hospital, Wilton, Cork, Ireland
| |
Collapse
|
16
|
Wang J, Wang Y, Jiang X. Targeting anticancer immunity in melanoma tumour microenvironment: unleashing the potential of adjuvants, drugs, and phytochemicals. J Drug Target 2024; 32:1052-1072. [PMID: 39041142 DOI: 10.1080/1061186x.2024.2384071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Melanoma poses a challenge in oncology because of its aggressive nature and limited treatment modalities. The tumour microenvironment (TME) in melanoma contains unique properties such as an immunosuppressive and high-density environment, unusual vasculature, and a high number of stromal and immunosuppressive cells. In recent years, numerous experiments have focused on boosting the immune system to effectively remove malignant cells. Adjuvants, consisting of phytochemicals, toll-like receptor (TLR) agonists, and cytokines, have shown encouraging results in triggering antitumor immunity and augmenting the therapeutic effectiveness of anticancer therapy. These adjuvants can stimulate the maturation of dendritic cells (DCs) and infiltration of cytotoxic CD8+ T lymphocytes (CTLs). Furthermore, nanocarriers can help to deliver immunomodulators and antigens directly to the tumour stroma, thereby improving their efficacy against malignant cells. The remodelling of melanoma TME utilising phytochemicals, agonists, and other adjuvants can be combined with current modalities for improving therapy outcomes. This review article explores the potential of adjuvants, drugs, and their nanoformulations in enhancing the anticancer potency of macrophages, CTLs, and natural killer (NK) cells. Additionally, the capacity of these agents to repress the function of immunosuppressive components of melanoma TME, such as immunosuppressive subsets of macrophages, stromal and myeloid cells will be discussed.
Collapse
Affiliation(s)
- Jingping Wang
- Emergency Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Yaping Wang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Xiaofang Jiang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| |
Collapse
|
17
|
Wang Y, Gao B, Jiao T, Zhang W, Shi H, Jiang H, Li X, Li J, Ge X, Pan K, Li C, Mao G, Lu S. CCL5/CCR5/CYP1A1 pathway prompts liver cancer cells to survive in the combination of targeted and immunological therapies. Cancer Sci 2024; 115:3552-3569. [PMID: 39183447 PMCID: PMC11531955 DOI: 10.1111/cas.16320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024] Open
Abstract
Combination therapy of anti-programmed cell death protein-1 (PD-1) antibodies and tyrosine kinase inhibitors (TKIs) has significantly improved the prognosis for hepatocellular carcinoma (HCC), but many patients still have unsatisfactory outcomes. CD8 T cells are known to exert a pivotal function in the immune response against tumors. Nevertheless, most CD8 T cells in HCC tissues are in a state of exhaustion, losing the cytotoxic activity against malignant cells. Cytokines, mainly secreted by immune cells, play an important role in the occurrence and development of tumors. Here, we demonstrated the changes in exhausted CD8T cells during combination therapy by single-cell RNA sequencing (scRNA-seq) analysis on tumor samples before and after treatment. Combination therapy exerted a substantial impact on the exhausted CD8T cells, particularly in terms of cytokine expression. CCL5 was the most abundantly expressed cytokine in CD8T cells and exhausted CD8T cells, and its expression increased further after treatment. Subsequently, we discovered the CCL5/CCR5/CYP1A1 pathway through RNA sequencing (RNA-seq) on CCL5-stimulated Huh7 cells and verified through a series of experiments that this pathway can mediate the resistance of liver cancer cells to lenvatinib. Tissue experiments showed that after combination therapy, the CCL5/CCR5/CYP1A1 pathway was activated, which can benefit the residual tumor cells to survive treatment. Tumor-bearing mouse experiments demonstrated that bergamottin (BGM), a competitive inhibitor of CYP1A1, can enhance the efficacy of both lenvatinib and combination therapy. Our research revealed one mechanism by which hepatoma cells can survive the combination therapy, providing a theoretical basis for the refined treatment of HCC.
Collapse
Affiliation(s)
- Yafei Wang
- Nankai University School of Medicine, Nankai UniversityTianjinChina
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Biao Gao
- Nankai University School of Medicine, Nankai UniversityTianjinChina
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Tianyu Jiao
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Wenwen Zhang
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Huizhong Shi
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Hao Jiang
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Xuerui Li
- Nankai University School of Medicine, Nankai UniversityTianjinChina
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Junfeng Li
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Xinlan Ge
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Ke Pan
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Chonghui Li
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Guankun Mao
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Shichun Lu
- Nankai University School of Medicine, Nankai UniversityTianjinChina
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| |
Collapse
|
18
|
Xiong N, Han W, Yu Z. ABO Blood Type and Pretreatment Systemic Inflammatory Response Index Associated with Lymph Node Metastasis in Patients with Breast Cancer. Int J Gen Med 2024; 17:4823-4833. [PMID: 39465189 PMCID: PMC11512788 DOI: 10.2147/ijgm.s486873] [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: 07/13/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024] Open
Abstract
Background Lymph node metastasis (LNM) is an important prognostic factor for breast cancer. Inflammatory stimulation can change tumor microenvironment and lead to LNM, but the relationship between LNM and peripheral immunoinflammatory indices has not been clarified in breast cancer. Methods The clinical information of 1918 patients with breast cancer admitted to Meizhou People's Hospital from October 2017 to December 2023 were retrospectively analyzed. The relationship of clinicopathological features (age, body mass index (BMI), ABO blood types, family history of cancer, tumor site, disease stage, LNM, distant metastasis, and molecular subtypes) and peripheral immunoinflammatory indices (pan-immune inflammation value (PIV), systemic immune inflammation index (SII), and system inflammation response index (SIRI)) were analyzed. Results There were 935 (48.7%) patients had no LNM and 983 (51.3%) had LNM. There were statistically significant differences in the distributions of ABO blood groups (p=0.022) and molecular subtypes (p<0.001) between the two groups. PIV, SII, and SIRI levels in patients with LNM were significantly higher than those without LNM (all p<0.05). The proportions of LNM in patients with high PIV, SII, and SIRI levels were higher than those with low PIV, SII, and SIRI levels, respectively. Logistic regression analysis showed that non-O blood type (non-O blood type vs O blood type, odds ratio (OR): 1.327, 95% confidence interval (CI): 1.056-1.667, p=0.015), luminal B subtype (luminal B vs luminal A, OR: 2.939, 95% CI: 2.147-4.022, p<0.001), HER2+ subtype (HER2+ vs luminal A, OR: 2.044, 95% CI: 1.388-3.009, p<0.001), and high SIRI level (≥0.875 vs <0.875, OR: 1.572, 95% CI: 1.092-2.265, p=0.015) were independently associated with LNM. Conclusion Non-O blood type, luminal B and HER2+ subtypes, and high SIRI level (≥0.875) have potential role in predicting the status of LNM in breast cancer patients.
Collapse
Affiliation(s)
- Nating Xiong
- Department of Blood Transfusion, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| | - Wendao Han
- Department of Blood Transfusion, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| | - Zhikang Yu
- Institute of Basic Medical Sciences, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
- Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| |
Collapse
|
19
|
Skapinker E, Aucoin EB, Kombargi HL, Yaish AM, Li Y, Baghaie L, Szewczuk MR. Contemporaneous Inflammatory, Angiogenic, Fibrogenic, and Angiostatic Cytokine Profiles of the Time-to-Tumor Development by Cancer Cells to Orchestrate Tumor Neovascularization, Progression, and Metastasis. Cells 2024; 13:1739. [PMID: 39451257 PMCID: PMC11506673 DOI: 10.3390/cells13201739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Cytokines can promote various cancer processes, such as angiogenesis, epithelial to mesenchymal transition (EMT), invasion, and tumor progression, and maintain cancer stem-cell-like (CSCs) cells. The mechanism(s) that continuously promote(s) tumors to progress in the TME still need(s) to be investigated. The data in the present study analyzed the inflammatory, angiogenic, fibrogenic, and angiostatic cytokine profiles in the host serum during tumor development in a mouse model of human pancreatic cancer. Pancreatic MiaPaCa-2-eGFP cancer cells were subcutaneously implanted in RAG2xCγ double mutant mice. Blood samples were collected before cancer cell implantation and every week until the end point of the study. The extracted serum from the blood of each mouse at different time points during tumor development was analyzed using a Bio-Plex microarray analysis and a Bio-Plex 200 system for proinflammatory (IL-1β, IL-10, IFN-γ, and TNF-α) and angiogenic and fibrogenic (IL-15, IL-18, basic FGF, LIF, M-CSF, MIG, MIP-2, PDGF-BB, and VEGF) cytokines. Here, we find that during cancer cell colonization for tumor development, host angiogenic, fibrogenic, and proinflammatory cytokine profiling in the tumor-bearing mice has been shown to significantly reduce host angiostatic and proinflammatory cytokines that restrain tumor development and increase those for tumor growth. The proinflammatory cytokines IL-15, IL-18, and IL-1β profiles reveal a significant host serum increase after day 35 when the tumor began to progress in growth. In contrast, the angiostatic cytokine profiles of TNFα, MIG, M-CSF, IL-10, and IFNγ in the host serum revealed a dramatic and significant decrease after day 5 post-implantation of cancer cells. OP treatment of tumor-bearing mice on day 35 maintained high levels of angiostatic and fibrogenic cytokines. The data suggest an entirely new regulation by cancer cells for tumor development. The findings identify for the first time how pancreatic cancer cells use host cytokine profiling to orchestrate the initiation of tumor development.
Collapse
Affiliation(s)
- Elizabeth Skapinker
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (E.S.); (Y.L.)
| | - Emilyn B. Aucoin
- Faculty of Science, Biology (Biomedical Science), York University, Toronto, ON M3J 1P3, Canada;
| | - Haley L. Kombargi
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada; (H.L.K.); (A.M.Y.)
| | - Abdulrahman M. Yaish
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada; (H.L.K.); (A.M.Y.)
| | - Yunfan Li
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (E.S.); (Y.L.)
| | - Leili Baghaie
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada;
| | - Myron R. Szewczuk
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada;
| |
Collapse
|
20
|
Heimes AS, Shehaj I, Almstedt K, Krajnak S, Schwab R, Stewen K, Lebrecht A, Brenner W, Hasenburg A, Schmidt M. Prognostic Impact of Acute and Chronic Inflammatory Interleukin Signatures in the Tumor Microenvironment of Early Breast Cancer. Int J Mol Sci 2024; 25:11114. [PMID: 39456897 PMCID: PMC11507514 DOI: 10.3390/ijms252011114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Interleukins play dual roles in breast cancer, acting as both promoters and inhibitors of tumorigenesis within the tumor microenvironment, shaped by their inflammatory functions. This study analyzed the subtype-specific prognostic significance of an acute inflammatory versus a chronic inflammatory interleukin signature using microarray-based gene expression analysis. Correlations between these interleukin signatures and immune cell markers (CD8, IgKC, and CD20) and immune checkpoints (PD-1) were also evaluated. This study investigated the prognostic significance of an acute inflammatory IL signature (IL-12, IL-21, and IFN-γ) and a chronic inflammatory IL signature (IL-4, IL-5, IL-10, IL-13, IL-17, and CXCL1) for metastasis-free survival (MFS) using Kaplan-Meier curves and Cox regression analyses in a cohort of 461 patients with early breast cancer. Correlations were analyzed using the Spearman-Rho correlation coefficient. Kaplan-Meier curves revealed that the prognostic significance of the acute inflammatory IL signature was specifically pronounced in the basal-like subtype (p = 0.004, Log Rank). This signature retained independent prognostic significance in multivariate Cox regression analysis (HR 0.463, 95% CI 0.290-0.741; p = 0.001). A higher expression of the acute inflammatory IL signature was associated with longer MFS. The chronic inflammatory IL signature showed a significant prognostic effect in the whole cohort, with higher expression associated with shorter MFS (p = 0.034). Strong correlations were found between the acute inflammatory IL signature and CD8 expression (ρ = 0.391; p < 0.001) and between the chronic inflammatory IL signature and PD-1 expression (ρ = 0.627; p < 0.001). This study highlights the complex interaction between acute and chronic inflammatory interleukins in breast cancer progression and prognosis. These findings provide insight into the prognostic relevance of interleukin expression patterns in breast cancer and may inform future therapeutic strategies targeting the immune-inflammatory axis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Marcus Schmidt
- Department of Obstetrics and Gynecology, University Medical Center of Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (A.-S.H.); (I.S.); (K.A.); (S.K.); (R.S.); (K.S.); (A.L.); (W.B.); (A.H.)
| |
Collapse
|
21
|
Pavlik T, Konchekov E, Shimanovskii N. Antitumor progestins activity: Cytostatic effect and immune response. Steroids 2024; 210:109474. [PMID: 39048056 DOI: 10.1016/j.steroids.2024.109474] [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: 04/25/2024] [Revised: 07/13/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
Progestins are used to treat some hormone-sensitive tumors. This review discusses the mechanisms of progestins' effects on tumor cells, the differences in the effects of progesterone and its analogs on different tumor types, and the influence of progestins on the antitumor immune response. Progestins cause a cytostatic effect, but at the same time they can suppress the antitumor immune response, and this can promote the proliferation and metastasis of tumor cells. Such progestins as dienogest, megestrol acetate and levonorgestrel increase the activity of NK-cells, which play a major role in the body's fight against tumor cells. The use of existing progestins and the development of new drugs with gestagenic activity may hold promise in oncotherapy.
Collapse
Affiliation(s)
- T Pavlik
- Pirogov Russian National Research Medical University, Russia; Prokhorov General Physics Institute of the Russian Academy of Sciences, Russia.
| | - E Konchekov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Russia; Peoples Friendship University of Russia (RUDN University), Russia
| | - N Shimanovskii
- Pirogov Russian National Research Medical University, Russia
| |
Collapse
|
22
|
Metwally YF, Elsaid AM, Elsadda RR, Refaat S, Zahran RF. Impact of IL-6 and IL-1β Gene Variants on Non-small-cell Lung Cancer Risk in Egyptian Patients. Biochem Genet 2024; 62:3367-3388. [PMID: 38103126 PMCID: PMC11427554 DOI: 10.1007/s10528-023-10596-2] [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: 07/26/2023] [Accepted: 11/08/2023] [Indexed: 12/17/2023]
Abstract
Lung cancer is a serious health and life issue, with the fastest-growing incidence and fatality rates worldwide. It is now clear that inflammation is a key factor involved in all aspects of carcinogenesis, notably lung cancer development. Genetic changes, including polymorphisms in inflammatory genes, are supposed to be a significant cause of increased lung cancer risk. The main idea of this research was to disclose the linkage between both IL-6 rs1800795 and IL-1β rs16944 variants and susceptibility to non-small-cell lung cancer (NSCLC) in Egyptians. This case-control design was composed of 127 cases and 138 controls, which were genotyped using the ARMS-PCR technique. To examine the NSCLC susceptibility under various genetic models, the odds ratio (OR) and 95% confidence intervals (CIs) were determined by logistic regression. Rs1800795 of the IL-6 gene was linked to higher odds of NSCLC under the allele model (adjusted, OR 2.28; 95% CI 1.2-4.33; p = 0.011). In the genetic models, IL-6 rs1800795 elevated the odds of NSCLC, while IL-1β rs16944 decreased the odds of NSCLC. Stratification analysis showed that IL-6 rs1800795 greatly increased the NSCLC risk in females and adenocarcinoma subtypes, whereas IL-1β rs16944 largely decreased the NSCLC risk for males, patients aged < 55, and nonsmokers. Regarding clinical data, the IL-6 variant was remarkably correlated with tumor size. This work primarily established that IL-6 and IL-1β variants have a great impact on NSCLC development in the Egyptian population; thus, it may be a supportive guide for earlier NSCLC prevention.
Collapse
Affiliation(s)
- Yomna F Metwally
- Biochemistry Department, Faculty of Science, Damietta University, Damietta, Egypt.
| | - Afaf M Elsaid
- Genetics Unit, Children Hospital, Mansoura University, Mansoura, Egypt
| | - Rana R Elsadda
- Biochemistry Department, Faculty of Science, Damietta University, Damietta, Egypt
| | - Sherif Refaat
- Oncology Department, Oncology Center, Mansoura University, Mansoura, Egypt
| | - Rasha F Zahran
- Biochemistry Department, Faculty of Science, Damietta University, Damietta, Egypt
| |
Collapse
|
23
|
Chen X, Peng H, Zhang Z, Yang C, Liu Y, Chen Y, Yu F, Wu S, Cao L. SPDYC serves as a prognostic biomarker related to lipid metabolism and the immune microenvironment in breast cancer. Immunol Res 2024; 72:1030-1050. [PMID: 38890248 DOI: 10.1007/s12026-024-09505-5] [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: 04/23/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Abstract
Breast cancer remains the most common malignant carcinoma among women globally and is resistant to several therapeutic agents. There is a need for novel targets to improve the prognosis of patients with breast cancer. Bioinformatics analyses were conducted to explore potentially relevant prognostic genes in breast cancer using The Cancer Genome Atlas (TCGA) and The Gene Expression Omnibus (GEO) databases. Gene subtypes were categorized by machine learning algorithms. The machine learning-related breast cancer (MLBC) score was evaluated through principal component analysis (PCA) of clinical patients' pathological statuses and subtypes. Immune cell infiltration was analyzed using the xCell and CIBERSORT algorithms. Kyoto Encyclopedia of Genes and Genomes enrichment analysis elucidated regulatory pathways related to speedy/RINGO cell cycle regulator family member C (SPDYC) in breast cancer. The biological functions and lipid metabolic status of breast cancer cell lines were validated via quantitative real-time polymerase chain reaction (RT‒qPCR) assays, western blotting, CCK-8 assays, PI‒Annexin V fluorescence staining, transwell assays, wound healing assays, and Oil Red O staining. Key differentially expressed genes (DEGs) in breast cancer from the TCGA and GEO databases were screened and utilized to establish the MLBC score. Moreover, the MLBC score we established was negatively correlated with poor prognosis in breast cancer patients. Furthermore, the impacts of SPDYC on the tumor immune microenvironment and lipid metabolism in breast cancer were revealed and validated. SPDYC is closely related to activated dendritic cells and macrophages and is simultaneously correlated with the immune checkpoints CD47, cytotoxic T lymphocyte antigen-4 (CTLA-4), and poliovirus receptor (PVR). SPDYC strongly correlated with C-C motif chemokine ligand 7 (CCL7), a chemokine that influences breast cancer patient prognosis. A significant relationship was discovered between key genes involved in lipid metabolism and SPDYC, such as ELOVL fatty acid elongase 2 (ELOVL2), malic enzyme 1 (ME1), and squalene epoxidase (SQLE). Potent inhibitors targeting SPDYC in breast cancer were also discovered, including JNK inhibitor VIII, AICAR, and JW-7-52-1. Downregulation of SPDYC expression in vitro decreased proliferation, increased the apoptotic rate, decreased migration, and reduced lipid droplets. SPDYC possibly influences the tumor immune microenvironment and regulates lipid metabolism in breast cancer. Hence, this study identified SPDYC as a pivotal biomarker for developing therapeutic strategies for breast cancer.
Collapse
Affiliation(s)
- Xinxin Chen
- Department of Breast Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haojie Peng
- Department of Breast Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhentao Zhang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Changnian Yang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yingqi Liu
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yanzhen Chen
- Department of Gynecology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Fei Yu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Shanshan Wu
- Department of Biology, School of Basic Medical Science, Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Lixue Cao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
24
|
Promny T, Scherrer I, Kadam S, Schmid R, Jost T, Distel LV, Arkudas A, Horch RE, Kengelbach-Weigand A. The Effect of Ionizing Irradiation on the Autotaxin-Lysophasphatidic Acid Axis and Interleukin-6/8 Secretion in Different Breast Cancer Cell Lines. J Pers Med 2024; 14:968. [PMID: 39338222 PMCID: PMC11433306 DOI: 10.3390/jpm14090968] [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: 07/30/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The Autotaxin (ATX)-lysophosphatidic acid (LPA) axis is involved in decreasing radiation sensitivity of breast tumor cells. This study aims to further elucidate the effect of irradiation on the ATX-LPA axis and cytokine secretion in different breast cancer cell lines to identify suitable breast cancer subtypes for targeted therapies. METHODS Different breast cancer cell lines (MCF-7 (luminal A), BT-474 (luminal B), SKBR-3 (HER2-positive), MDA-MB-231 and MDA-MB-468 (triple-negative)) and the breast epithelial cell line MCF-10A were irradiated. The influence of irradiation on LPA receptor (LPAR) expression, ATX expression, and Interleukin (IL)-6 and IL-8 secretion was analyzed. Further, the effect of IL-6 and IL-8 on ATX expression of adipose-derived stem cells (ADSC) was investigated. RESULTS Irradiation increased ATX and LPAR2 expression in MDA-MB-231 cells. Additionally, IL-6 secretion was enhanced in MDA-MB-231, and IL-8 secretion in MDA-MB-231 and MDA-MB-468. Stimulation of ADSC with IL-6 and IL-8 increased ATX expression in ADSC. CONCLUSIONS Targeting ATX or its downstream signaling pathways might enhance the sensitivity of triple-negative breast cancer cells to radiation. Further exploration of the interplay between irradiation, the ATX-LPA axis, and inflammatory cytokines may elucidate novel pathways for overcoming radioresistance and improving individual treatment outcomes.
Collapse
Affiliation(s)
- Theresa Promny
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Isabell Scherrer
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Sheetal Kadam
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Rafael Schmid
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Tina Jost
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Luitpold V Distel
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Andreas Arkudas
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Raymund E Horch
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Annika Kengelbach-Weigand
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
25
|
Tirgar A, Rezaei M, Ehsani M, Salmani Z, Rastegari A, Jafari E, Khandani BK, Nakhaee N, Khaksari M, Moazed V. Exploring the synergistic effects of vitamin D and synbiotics on cytokines profile, and treatment response in breast cancer: a pilot randomized clinical trial. Sci Rep 2024; 14:21372. [PMID: 39266591 PMCID: PMC11393349 DOI: 10.1038/s41598-024-72172-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 09/04/2024] [Indexed: 09/14/2024] Open
Abstract
This study was designed to investigate the effect of vitamin D and/or synbiotics on the response to treatment, cytokines profile and hormonal biomarkers in breast cancer patients undergoing neoadjuvant therapy. A total of 76 patients were recruited and completed the course of the intervention between 2019 and 2021 in Kerman, Iran. breast cancer patients were randomly enrolled in this study. Patients divided into four groups to receive one of the following regimens: placebo, vitamin D, synbiotics and a combination of vitamin D and synbiotics. clinicopathologic parameters, inflammatory and anti-inflammatory biomarkers and hormonal levels were measured at the baseline and four months after intervention. The study results found no clear link between the interventions and achieving pathological complete response (pCR), and a similar trend was observed in Ki-67 index examination. After neoadjuvant therapy, TNF-α concentrations decreased, with vitamin D supplementation moderating this decline. Vitamin D supplemented groups showed a significant increase in serum IL-6 levels. While IL-10 levels decreased in the placebo group, all intervention groups were protected from this decline. Moreover, there was a notable increase in the anti-inflammatory index, particularly in the group receiving both vitamin D and synbiotic supplementation, suggesting potential synergistic anti-inflammatory effects from their combined administration. The outcomes suggest a potential anti-inflammatory function of this combination. Consequently, more extensive studies with prolonged follow-up periods and substantial sample sizes are warranted to thoroughly evaluate their potential benefits for breast cancer patients.
Collapse
Grants
- 98000252 Kerman University of Medical Sciences, Iran
- 98000252 Kerman University of Medical Sciences, Iran
- 98000252 Kerman University of Medical Sciences, Iran
- 98000252 Kerman University of Medical Sciences, Iran
- 98000252 Kerman University of Medical Sciences, Iran
- 98000252 Kerman University of Medical Sciences, Iran
- 98000252 Kerman University of Medical Sciences, Iran
- 98000252 Kerman University of Medical Sciences, Iran
- 98000252 Kerman University of Medical Sciences, Iran
- 98000252 Kerman University of Medical Sciences, Iran
Collapse
Affiliation(s)
- Aliasghar Tirgar
- Department of Hematology and Oncology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Masoud Rezaei
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Zahra Salmani
- Department of Hematology and Oncology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Armin Rastegari
- Department of Hematology and Oncology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Jafari
- Department of Pathology, Pathology and Stem Cell Research Center, School of Medicine, Kerman University of Medical Science, Kerman, Iran
| | - Behjat Kalantari Khandani
- Department of Hematology and Oncology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Nouzar Nakhaee
- Health Services Management Research Center, Institute of Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Department of Physiology and Pharmacology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Vahid Moazed
- Department of Hematology and Oncology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
26
|
Guo J, Zhao J, Tian P, Xu Z, Wang R, Chen W, Wang X, Wan S, Yang Y, Zhang H. BaP/BPDE exposure causes human trophoblast cell dysfunctions and induces miscarriage by up-regulating lnc-HZ06-regulated IL1B. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:134741. [PMID: 38991640 DOI: 10.1016/j.jhazmat.2024.134741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/11/2024] [Accepted: 05/24/2024] [Indexed: 07/13/2024]
Abstract
Exposure to environmental BaP or its metabolite BPDE causes trophoblast cell dysfunctions to induce miscarriage (abnormal early embryo loss), which might be generally regulated by lncRNAs. IL1B, a critical inflammatory cytokine, is closely associated with adverse pregnancy outcomes. However, whether IL1B might cause dysfunctions of BaP/BPDE-exposed trophoblast cells to induce miscarriage, as well as its specific epigenetic regulatory mechanisms, is completely unexplored. In this study, we find that BPDE-DNA adducts, trophoblast cell dysfunctions, and miscarriage are closely associated. Moreover, we also identify a novel lnc-HZ06 and IL1B, both of which are highly expressed in BPDE-exposed trophoblast cells, in villous tissues of recurrent miscarriage patients, and in placental tissues of BaP-exposed mice with miscarriage. Both lnc-HZ06 and IL1B suppress trophoblast cell migration/invasion and increase apoptosis. In mechanism, lnc-HZ06 promotes STAT4-mediated IL1B mRNA transcription, enhances IL1B mRNA stability by promoting the formation of METTL3/HuR/IL1B mRNA ternary complex, and finally up-regulates IL1B expression levels. BPDE exposure promotes TBP-mediated lnc-HZ06 transcription, and thus up-regulates IL1B levels. Knockdown of either murine lnc-hz06 (which down-regulates Il1b levels) or murine Il1b could alleviate miscarriage in BaP-exposed mice. Collectively, this study not only discovers novel biological mechanisms and pathogenesis of unexplained miscarriage but also provides novel potential targets for treatment against BaP/BPDE-induced miscarriage.
Collapse
Affiliation(s)
- Jiarong Guo
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China; Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu 610041, China; Chengdu Qingyang District for Disease Control and Prevention, Chengdu 610031, China
| | - Jingsong Zhao
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China; Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Peng Tian
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China; Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Zhongyan Xu
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Rong Wang
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Weina Chen
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Xiaoqing Wang
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Shukun Wan
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Yang Yang
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Huidong Zhang
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China.
| |
Collapse
|
27
|
Zhang Y, Ding X, Zhang X, Li Y, Xu R, Li HJ, Zuo D, Chen G. Unveiling the contribution of tumor-associated macrophages in driving epithelial-mesenchymal transition: a review of mechanisms and therapeutic Strategies. Front Pharmacol 2024; 15:1404687. [PMID: 39286635 PMCID: PMC11402718 DOI: 10.3389/fphar.2024.1404687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 08/15/2024] [Indexed: 09/19/2024] Open
Abstract
Tumor-associated macrophages (TAMs), fundamental constituents of the tumor microenvironment (TME), significantly influence cancer development, primarily by promoting epithelial-mesenchymal transition (EMT). EMT endows cancer cells with increased motility, invasiveness, and resistance to therapies, marking a pivotal juncture in cancer progression. The review begins with a detailed exposition on the origins of TAMs and their functional heterogeneity, providing a foundational understanding of TAM characteristics. Next, it delves into the specific molecular mechanisms through which TAMs induce EMT, including cytokines, chemokines and stromal cross-talking. Following this, the review explores TAM-induced EMT features in select cancer types with notable EMT characteristics, highlighting recent insights and the impact of TAMs on cancer progression. Finally, the review concludes with a discussion of potential therapeutic targets and strategies aimed at mitigating TAM infiltration and disrupting the EMT signaling network, thereby underscoring the potential of emerging treatments to combat TAM-mediated EMT in cancer. This comprehensive analysis reaffirms the necessity for continued exploration into TAMs' regulatory roles within cancer biology to refine therapeutic approaches and improve patient outcomes.
Collapse
Affiliation(s)
- Yijia Zhang
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaofei Ding
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang, China
| | - Xue Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Ye Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Rui Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Hai-Jun Li
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Guang Chen
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang, China
| |
Collapse
|
28
|
Wang Y, Li D, Li Q, Basnet A, Efird JT, Seki N. Neutrophil estimation and prognosis analysis based on existing lung squamous cell carcinoma datasets: the development and validation of a prognosis prediction model. Transl Lung Cancer Res 2024; 13:2023-2037. [PMID: 39263021 PMCID: PMC11384491 DOI: 10.21037/tlcr-24-411] [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: 05/08/2024] [Accepted: 08/06/2024] [Indexed: 09/13/2024]
Abstract
Background Notwithstanding the rapid developments in precision medicine in recent years, lung cancer still has a low survival rate, especially lung squamous cell cancer (LUSC). The tumor microenvironment (TME) plays an important role in the progression of lung cancer, in which high neutrophil levels are correlated with poor prognosis, potentially due to their interactions with tumor cells via pro-inflammatory cytokines and chemokines. However, the precise mechanisms of how neutrophils influence lung cancer remain unclear. This study aims to explore these mechanisms and develop a prognosis predictive model in LUSC, addressing the knowledge gap in neutrophil-related cancer pathogenesis. Methods LUSC datasets from the Xena Hub and Gene Expression Omnibus (GEO) databases were used, comprising 473 tumor samples and 195 tumor samples, respectively. Neutrophil contents in these samples were estimated using CIBERSORT, xCell, and microenvironment cell populations (MCP) counter tools. Differentially expressed genes (DEGs) were identified using DEseq2, and a weighted gene co-expression network analysis (WGCNA) was performed to identify neutrophil-related genes. A least absolute shrinkage and selection operator (LASSO) Cox regression model was constructed for prognosis prediction, and the model's accuracy was validated using Kaplan-Meier survival curves and time-dependent receiver operating characteristic (ROC) curves. Additionally, genomic changes, immune correlations, drug sensitivity, and immunotherapy response were analyzed to further validate the model's predictive power. Results Neutrophil content was significantly higher in adjacent normal tissue compared to LUSC tissue (P<0.001). High neutrophil content was associated with worse overall survival (OS) (P=0.02), disease-free survival (DFS) (P=0.02), and progression-free survival (PFS) (P=0.03) using different software estimates. Nine gene modules were identified, with blue and yellow modules showing strong correlations with neutrophil prognosis (P<0.001). Eight genes were selected for the prognostic model, which accurately predicted 1-, 3-, and 5-year survival in both the training set [area under the curve (AUC) value =0.60, 0.63, 0.66, respectively] and validation set (AUC value =0.58, 0.58, 0.59, respectively), with significant prognosis differences between high- and low-risk groups (P<0.001). The model's independent prognostic factors included risk group, pathologic M stage, and tumor stage (P<0.05). A further molecular mechanism analysis revealed differences between risk groups were revealed in immune checkpoint and human leukocyte antigen (HLA) gene expression, hallmark pathways, drug sensitivity, and immunotherapy responses. Conclusions This study established a risk-score model that effectively predicts the prognosis of LUSC patients and sheds light on the molecular mechanisms involved. The findings enhance the understanding of neutrophil-tumor interactions, offering potential targets for personalized treatments. However, further experimental validation and clinical studies are required to confirm these findings and address study limitations, including reliance on public databases and focus on a specific lung cancer subtype.
Collapse
Affiliation(s)
- Youyu Wang
- Department of Thoracic Surgery, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Dongfang Li
- Department of Thoracic Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Qiang Li
- Department of Oncology, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Alina Basnet
- Division of Hematology-Oncology, Upstate Cancer Center, Upstate Medical University, Syracuse, NY, USA
| | - Jimmy T Efird
- VA Cooperative Studies Program Coordinating Center, Boston, MA, USA
- Department of Radiation Oncology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Nobuhiko Seki
- Division of Medical Oncology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
29
|
Tomecka P, Kunachowicz D, Górczyńska J, Gebuza M, Kuźnicki J, Skinderowicz K, Choromańska A. Factors Determining Epithelial-Mesenchymal Transition in Cancer Progression. Int J Mol Sci 2024; 25:8972. [PMID: 39201656 PMCID: PMC11354349 DOI: 10.3390/ijms25168972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 09/02/2024] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a process in which an epithelial cell undergoes multiple modifications, acquiring both morphological and functional characteristics of a mesenchymal cell. This dynamic process is initiated by various inducing signals that activate numerous signaling pathways, leading to the stimulation of transcription factors. EMT plays a significant role in cancer progression, such as metastasis and tumor heterogeneity, as well as in drug resistance. In this article, we studied molecular mechanisms, epigenetic regulation, and cellular plasticity of EMT, as well as microenvironmental factors influencing this process. We included both in vivo and in vitro models in EMT investigation and clinical implications of EMT, such as the use of EMT in curing oncological patients and targeting its use in therapies. Additionally, this review concludes with future directions and challenges in the wide field of EMT.
Collapse
Affiliation(s)
- Paulina Tomecka
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (P.T.); (J.G.); (M.G.); (J.K.); (K.S.)
| | - Dominika Kunachowicz
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland;
| | - Julia Górczyńska
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (P.T.); (J.G.); (M.G.); (J.K.); (K.S.)
| | - Michał Gebuza
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (P.T.); (J.G.); (M.G.); (J.K.); (K.S.)
| | - Jacek Kuźnicki
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (P.T.); (J.G.); (M.G.); (J.K.); (K.S.)
| | - Katarzyna Skinderowicz
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (P.T.); (J.G.); (M.G.); (J.K.); (K.S.)
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland
| |
Collapse
|
30
|
Sun M, Bai J, Wang H, Li M, Zhou L, Li S. Unraveling the relationship between anoikis-related genes and cancer-associated fibroblasts in liver hepatocellular carcinoma. Heliyon 2024; 10:e35306. [PMID: 39165997 PMCID: PMC11334810 DOI: 10.1016/j.heliyon.2024.e35306] [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: 11/28/2023] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
This study intended to determine the molecular subtypes of liver hepatocellular carcinoma (LIHC) on the strength of anoikis-related genes (ARGs) and to assess their prognostic value and prospective relationship with immune cell infiltration and cancer-associated fibroblasts (CAFs). Univariate Cox regression analysis yielded 66 prognosis-related ARGs and classified LIHC into two distinct subtypes, with subtype A demonstrating overexpression of most prognosis-related ARGs and a significant survival disadvantage. Furthermore, a reliable prediction model was developed using ARGs to evaluate the risk of LIHC patients. This model served as an independent prognostic indicator and a quantitative tool for clinical prognostic prediction. Additionally, subtype-specific differences in immune cell infiltration were observed, and the risk score was potentially linked to immune-related characteristics. Moreover, the study identified a significant association between CAF score and LIHC prognosis, with a low CAF score indicating a favorable patient prognosis. In conclusion, this study reveals the molecular mechanisms underlying the development and progression of LIHC and identifies potential therapeutic targets for the disease.
Collapse
Affiliation(s)
- Meng Sun
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Jiangtao Bai
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Haisong Wang
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Mei Li
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Long Zhou
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Shanfeng Li
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
31
|
Jiang P, Zhu X, Jiang Y, Li H, Luo Q. Targeting JUNB to modulate M2 macrophage polarization in preeclampsia. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167194. [PMID: 38663490 DOI: 10.1016/j.bbadis.2024.167194] [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: 11/11/2023] [Revised: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 06/17/2024]
Abstract
Preeclampsia (PE) is a complex disorder affecting pregnant women, leading to significant maternal and fetal morbidity and mortality. Understanding the cellular dynamics and molecular mechanisms underlying PE is crucial for developing effective therapeutic strategies. This study utilized single-cell RNA sequencing (scRNA-seq) to delineate the cellular landscape of the placenta in PE, identifying 11 distinct cell subpopulations, with macrophages playing a pivotal role in mediating cell-cell communication. Specifically, the transcription factor JUNB was found to be a key gene in macrophages from PE samples, influencing the interaction between macrophages and both epithelial and endothelial cells. Functional experiments indicated that interference with JUNB expression promoted macrophage polarization towards an M2 phenotype, which facilitated trophoblast invasion, migration, and angiogenesis. Mechanistically, JUNB regulated the MIIP/PI3K/AKT pathway, as evidenced by gene expression analysis following JUNB knockdown. The study further demonstrated that targeting JUNB could activate the PI3K/AKT pathway by transcriptionally activating MIIP, thus promoting M2 polarization and potentially delaying the onset of PE. These findings present new insights into the pathogenesis of PE and suggest a novel therapeutic approach by modulating macrophage polarization.
Collapse
Affiliation(s)
- Peiyue Jiang
- Department of Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, PR China
| | - Xiaojun Zhu
- Department of Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, PR China
| | - Ying Jiang
- Department of Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, PR China
| | - Hetong Li
- Department of Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, PR China
| | - Qiong Luo
- Department of Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, PR China.
| |
Collapse
|
32
|
Almalki WH, Almujri SS. Circular RNAs and the JAK/STAT pathway: New frontiers in cancer therapeutics. Pathol Res Pract 2024; 260:155408. [PMID: 38909403 DOI: 10.1016/j.prp.2024.155408] [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: 05/01/2024] [Revised: 06/08/2024] [Accepted: 06/12/2024] [Indexed: 06/25/2024]
Abstract
Circular RNAs, known as circRNAs, have drawn more attention to cancer biology in the last few years. Novel functions of circRNAs in cancer therapy open promising prospects for personalized medicine. This review focuses on the molecular properties and potential of circRNAs as biomarkers or therapeutic targets in cancer treatment. Unique properties of circular RNAs associated with a circular form provide stability and resilience to RNA exonuclease degradation. Circular RNAs' most important characteristic is that they are involved in the JAK/STAT pathway associated with oncogenesis. Notably, their deregulation has been reported in multiple carcinomas due to involvement in JAK/STAT signaling cascade modulation. Increased knowledge about circRNAs' interaction with the JAK/STAT pathway leads to the emergence of new possibilities for targeted cancer therapy. In addition, since circRNAs demonstrate tissue-relatedness of expression, they may be a reliable biomarker for predicting and diagnosing cancer. With the development of new technologies for targeting circRNAs, novel therapeutics can be produced that offer more personalized cancer treatment options based on the nature of the patient. The present review explores the exciting prospects of circRNAs for transforming cancer treatment into personalized medicine. It describes the current understanding of circRNA biology, its relationship to tumorigenesis, and possible targeting methods.
Collapse
Affiliation(s)
- Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Aseer 61421, Saudi Arabia
| |
Collapse
|
33
|
Mao J, Liu L, Shen Q, Cen M. Integrating single-cell transcriptomics and machine learning to predict breast cancer prognosis: A study based on natural killer cell-related genes. J Cell Mol Med 2024; 28:e18549. [PMID: 39098994 PMCID: PMC11298315 DOI: 10.1111/jcmm.18549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/24/2024] [Accepted: 07/09/2024] [Indexed: 08/06/2024] Open
Abstract
Breast cancer (BC) is the most commonly diagnosed cancer in women globally. Natural killer (NK) cells play a vital role in tumour immunosurveillance. This study aimed to establish a prognostic model using NK cell-related genes (NKRGs) by integrating single-cell transcriptomic data with machine learning. We identified 44 significantly expressed NKRGs involved in cytokine and T cell-related functions. Using 101 machine learning algorithms, the Lasso + RSF model showed the highest predictive accuracy with nine key NKRGs. We explored cell-to-cell communication using CellChat, assessed immune-related pathways and tumour microenvironment with gene set variation analysis and ssGSEA, and observed immune components by HE staining. Additionally, drug activity predictions identified potential therapies, and gene expression validation through immunohistochemistry and RNA-seq confirmed the clinical applicability of NKRGs. The nomogram showed high concordance between predicted and actual survival, linking higher tumour purity and risk scores to a reduced immune score. This NKRG-based model offers a novel approach for risk assessment and personalized treatment in BC, enhancing the potential of precision medicine.
Collapse
Affiliation(s)
- Juanjuan Mao
- Department of Thyroid and Breast SurgeryNingbo Hospital of TCM Affiliated to Zhejiang Chinese Medicine UniversityNingbo CityZhejiang ProvinceChina
| | - Ling‐lin Liu
- Department of Thyroid and Breast SurgeryNingbo Hospital of TCM Affiliated to Zhejiang Chinese Medicine UniversityNingbo CityZhejiang ProvinceChina
| | - Qian Shen
- Department of Thyroid and Breast SurgeryNingbo Hospital of TCM Affiliated to Zhejiang Chinese Medicine UniversityNingbo CityZhejiang ProvinceChina
| | - Mengyan Cen
- Department of Thyroid and Breast SurgeryNingbo Hospital of TCM Affiliated to Zhejiang Chinese Medicine UniversityNingbo CityZhejiang ProvinceChina
| |
Collapse
|
34
|
Miller ZA, Muthuswami S, Mueller A, Ma RZ, Sywanycz SM, Naik A, Huang L, Brody RM, Diab A, Carey RM, Lee RJ. GLUT1 inhibitor BAY-876 induces apoptosis and enhances anti-cancer effects of bitter receptor agonists in head and neck squamous carcinoma cells. Cell Death Discov 2024; 10:339. [PMID: 39060287 PMCID: PMC11282258 DOI: 10.1038/s41420-024-02106-z] [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: 03/27/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are cancers that arise in the mucosa of the upper aerodigestive tract. The five-year patient survival rate is ~50%. Treatment includes surgery, radiation, and/or chemotherapy and is associated with lasting effects even when successful in irradicating the disease. New molecular targets and therapies must be identified to improve outcomes for HNSCC patients. We recently identified bitter taste receptors (taste family 2 receptors, or T2Rs) as a novel candidate family of receptors that activate apoptosis in HNSCC cells through mitochondrial Ca2+ overload and depolarization. We hypothesized that targeting another component of tumor cell metabolism, namely glycolysis, may increase the efficacy of T2R-directed therapies. GLUT1 (SLC2A1) is a facilitated-diffusion glucose transporter expressed by many cancer cells to fuel their increased rates of glycolysis. GLUT1 is already being investigated as a possible cancer target, but studies in HNSCCs are limited. Examination of immortalized HNSCC cells, patient samples, and The Cancer Genome Atlas revealed high expression of GLUT1 and upregulation in some patient tumor samples. HNSCC cells and tumor tissue express GLUT1 on the plasma membrane and within the cytoplasm (perinuclear, likely co-localized with the Golgi apparatus). We investigated the effects of a recently developed small molecule inhibitor of GLUT1, BAY-876. This compound decreased HNSCC glucose uptake, viability, and metabolism and induced apoptosis. Moreover, BAY-876 had enhanced effects on apoptosis when combined at low concentrations with T2R bitter taste receptor agonists. Notably, BAY-876 also decreased TNFα-induced IL-8 production, indicating an additional mechanism of possible tumor-suppressive effects. Our study demonstrates that targeting GLUT1 via BAY-876 to kill HNSCC cells, particularly in combination with T2R agonists, is a potential novel treatment strategy worth exploring further in future translational studies.
Collapse
Affiliation(s)
- Zoey A Miller
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Pharmacology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Sahil Muthuswami
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Arielle Mueller
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Ray Z Ma
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Sarah M Sywanycz
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Anusha Naik
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Lily Huang
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Robert M Brody
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Ahmed Diab
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Ryan M Carey
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| | - Robert J Lee
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
35
|
Xue D, Zuo Q, Chang J, Wu X. The correlation between TRIM28 expression and immune checkpoints in CRPC. FASEB J 2024; 38:e23663. [PMID: 38958986 DOI: 10.1096/fj.202400061rr] [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: 01/09/2024] [Revised: 04/17/2024] [Accepted: 04/29/2024] [Indexed: 07/04/2024]
Abstract
This study delves into the unexplored realm of castration-resistant prostate cancer (CRPC) by investigating the role of TRIM28 and its intricate molecular mechanisms using high-throughput single-cell transcriptome sequencing and advanced bioinformatics analysis. Our comprehensive examination unveiled dynamic TRIM28 expression changes, particularly in immune cells such as macrophages and CD8+ T cells within CRPC. Correlation analyses with TCGA data highlighted the connection between TRIM28 and immune checkpoint expression and emphasized its pivotal influence on the quantity and functionality of immune cells. Using TRIM28 knockout mouse models, we identified differentially expressed genes and enriched pathways, unraveling the potential regulatory involvement of TRIM28 in the cGAS-STING pathway. In vitro, experiments further illuminated that TRIM28 knockout in prostate cancer cells induced a notable anti-tumor immune effect by inhibiting M2 macrophage polarization and enhancing CD8+ T cell activity. This impactful discovery was validated in an in situ transplant tumor model, where TRIM28 knockout exhibited a deceleration in tumor growth, reduced proportions of M2 macrophages, and enhanced infiltration of CD8+ T cells. In summary, this study elucidates the hitherto unknown anti-tumor immune role of TRIM28 in CRPC and unravels its potential regulatory mechanism via the cGAS-STING signaling pathway. These findings provide novel insights into the immune landscape of CRPC, offering promising directions for developing innovative therapeutic strategies.
Collapse
Affiliation(s)
- Dun Xue
- Department of Medical, the First Hospital of Changsha, Changsha, P. R. China
| | - Qian Zuo
- Department of Radiology, the First Hospital of Changsha, Changsha, P. R. China
| | - Jie Chang
- Department of Outpatient, the First Hospital of Changsha, Changsha, P. R. China
| | - Xinghui Wu
- Department of Urology, the First Hospital of Changsha, Changsha, P. R. China
| |
Collapse
|
36
|
Alharbi M, Roy AM, Krishnan J, Kalinski P, Yao S, Gandhi S. Targeting the tumor microenvironment to improve clinical outcomes in triple negative breast cancer patients and bridge the current disparity gap. Front Immunol 2024; 15:1428118. [PMID: 39072334 PMCID: PMC11272470 DOI: 10.3389/fimmu.2024.1428118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Triple negative breast cancer (TNBC) is a heterogenous disease that disproportionately affects Black women. TNBC outcomes among Black women are dismal secondary to multiple factors, such as poor healthcare accessibility resulting in delays in diagnosis, and aggressive disease biology in addition to a pro-tumor immune microenvironment (TME). Black women with breast cancer exhibit elevated levels of serum pro-inflammatory cytokines, and a pro-tumorigenic TME with higher immunosuppressive regulatory T cells (Tregs), M2 macrophages and exhausted CD8+ T cells. We have shown that the combined use of toll-like receptor 3 (TLR3) ligands with interferon-α (chemokine modulation: CKM) is able to enrich the tumor with CD8+ T cells, while not increasing immunosuppressive cells. Recent clinical trials have revealed the efficacy of immune checkpoint inhibitors (ICI) in rejuvenizing exhausted CD8+ T cells. We hypothesize that strategies to modulate the TME by enriching chemokines that attract CD8+T cells followed by reversal of CD8+ T cell exhaustion (ICI), when added to standard treatment, could potentially improve clinical outcomes, and mitigate the racial disparities in TNBC outcomes between Black and White Women.
Collapse
Affiliation(s)
- Malak Alharbi
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- Department of Internal Medicine, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Arya Mariam Roy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Jayasree Krishnan
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Pawel Kalinski
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Song Yao
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Shipra Gandhi
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
37
|
Habanjar O, Nehme R, Goncalves-Mendes N, Cueff G, Blavignac C, Aoun J, Decombat C, Auxenfans C, Diab-Assaf M, Caldefie-Chézet F, Delort L. The obese inflammatory microenvironment may promote breast DCIS progression. Front Immunol 2024; 15:1384354. [PMID: 39072314 PMCID: PMC11272476 DOI: 10.3389/fimmu.2024.1384354] [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: 02/09/2024] [Accepted: 06/20/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction Ductal carcinoma in situ (DCIS), characterized by a proliferation of neoplastic cells confined within the mammary ducts, is distinctly isolated from the surrounding stroma by an almost uninterrupted layer of myoepithelial cells (MECs) and by the basement membrane. Heightened interactions within the adipose microenvironment, particularly in obese patients, may play a key role in the transition from DCIS to invasive ductal carcinoma (IDC), which is attracting growing interest in scientific research. Adipose tissue undergoes metabolic changes in obesity, impacting adipokine secretion and promoting chronic inflammation. This study aimed to assess the interactions between DCIS, including in situ cancer cells and MECs, and the various components of its inflammatory adipose microenvironment (adipocytes and macrophages). Methods To this end, a 3D co-culture model was developed using bicellular bi-fluorescent DCIS-like tumoroids, adipose cells, and macrophages to investigate the influence of the inflammatory adipose microenvironment on DCIS progression. Results The 3D co-culture model demonstrated an inhibition of the expression of genes involved in apoptosis (BAX, BAG1, BCL2, CASP3, CASP8, and CASP9), and an increase in genes related to cell survival (TP53, JUN, and TGFB1), inflammation (TNF-α, PTGS2, IL-6R), invasion and metastasis (TIMP1 and MMP-9) in cancer cells of the tumoroids under inflammatory conditions versus a non-inflammatory microenvironment. On the contrary, it confirmed the compromised functionality of MECs, resulting in the loss of their protective effects against cancer cells. Adipocytes from obese women showed a significant increase in the expression of all studied myofibroblast-associated genes (myoCAFs), such as FAP and α-SMA. In contrast, adipocytes from normal-weight women expressed markers of inflammatory fibroblast phenotypes (iCAF) characterized by a significant increase in the expression of LIF and inflammatory cytokines such as TNF-α, IL-1β, IL-8, and CXCL-10. These changes also influenced macrophage polarization, leading to a pro-inflammatory M1 phenotype. In contrast, myoCAF-associated adipocytes, and the cancer-promoting microenvironment polarized macrophages towards an M2 phenotype, characterized by high CD163 receptor expression and IL-10 and TGF-β secretion. Discussion Reciprocal interactions between the tumoroid and its microenvironment, particularly in obesity, led to transcriptomic changes in adipocytes and macrophages, may participate in breast cancer progression while disrupting the integrity of the MEC layer. These results underlined the importance of adipose tissue in cancer progression.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | - Rawan Nehme
- Université Clermont-Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | | | - Gwendal Cueff
- Université Clermont-Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | - Christelle Blavignac
- Université Clermont-Auvergne, Centre d’Imagerie Cellulaire Santé (CCIS), Clermont-Ferrand, France
| | - Jessy Aoun
- Université Clermont-Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | | | - Céline Auxenfans
- Banque de tissus et de cellules, Hôpital Edouard-Herriot, Lyon, France
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Moléculaire et Pharmacologie Anticancéreuse, Faculté des Sciences II, Université libanaise Fanar, Beirut, Lebanon
| | | | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Clermont-Ferrand, France
| |
Collapse
|
38
|
Gerashchenko T, Frolova A, Patysheva M, Fedorov A, Stakheyeva M, Denisov E, Cherdyntseva N. Breast Cancer Immune Landscape: Interplay Between Systemic and Local Immunity. Adv Biol (Weinh) 2024; 8:e2400140. [PMID: 38727796 DOI: 10.1002/adbi.202400140] [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: 03/12/2024] [Revised: 04/16/2024] [Indexed: 07/13/2024]
Abstract
Breast cancer (BC) is one of the most common malignancies in women worldwide. Numerous studies in immuno-oncology and successful trials of immunotherapy have demonstrated the causal role of the immune system in cancer pathogenesis. The interaction between the tumor and the immune system is known to have a dual nature. Despite cytotoxic lymphocyte activity against transformed cells, a tumor can escape immune surveillance and leverage chronic inflammation to maintain its own development. Research on antitumor immunity primarily focuses on the role of the tumor microenvironment, whereas the systemic immune response beyond the tumor site is described less thoroughly. Here, a comprehensive review of the formation of the immune profile in breast cancer patients is offered. The interplay between systemic and local immune reactions as self-sustaining mechanism of tumor progression is described and the functional activity of the main cell populations related to innate and adaptive immunity is discussed. Additionally, the interaction between different functional levels of the immune system and their contribution to the development of the pro- or anti-tumor immune response in BC is highlighted. The presented data can potentially inform the development of new immunotherapy strategies in the treatment of patients with BC.
Collapse
Affiliation(s)
- Tatiana Gerashchenko
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Anastasia Frolova
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Researc, Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
- Tomsk State University, 36 Lenin Ave., Tomsk, 634050, Russia
| | - Marina Patysheva
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Anton Fedorov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Marina Stakheyeva
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Researc, Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Evgeny Denisov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Nadezda Cherdyntseva
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Researc, Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
- Tomsk State University, 36 Lenin Ave., Tomsk, 634050, Russia
| |
Collapse
|
39
|
Kumar DR, Banaś A, Krukiewicz K. Challenges and Advances in Biomarker Detection for Rapid and Accurate Sepsis Diagnosis: An Electrochemical Approach. BIOSENSORS 2024; 14:309. [PMID: 38920613 PMCID: PMC11202072 DOI: 10.3390/bios14060309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024]
Abstract
Sepsis is a life-threatening condition with high mortality rates due to delayed treatment of patients. The conventional methodology for blood diagnosis takes several hours, which suspends treatment, limits early drug administration, and affects the patient's recovery. Thus, rapid, accurate, bedside (onsite), economical, and reliable sepsis biomarker reading of the clinical sample is an emergent need for patient lifesaving. Electrochemical label-free biosensors are specific and rapid devices that are able to perform analysis at the patient's bedside; thus, they are considered an attractive methodology in a clinical setting. To reveal their full diagnostic potential, electrode architecture strategies of fabrication are highly desirable, particularly those able to preserve specific antibody-antigen attraction, restrict non-specific adsorption, and exhibit high sensitivity with a low detection limit for a target biomarker. The aim of this review is to provide state-of-the-art methodologies allowing the fabrication of ultrasensitive and highly selective electrochemical sensors for sepsis biomarkers. This review focuses on different methods of label-free biomarker sensors and discusses their advantages and disadvantages. Then, it highlights effective ways of avoiding false results and the role of molecular labels and functionalization. Recent literature on electrode materials and antibody grafting strategies is discussed, and the most efficient methodology for overcoming the non-specific attraction issues is listed. Finally, we discuss the existing electrode architecture for specific biomarker readers and promising tactics for achieving quick and low detection limits for sepsis biomarkers.
Collapse
Affiliation(s)
- Deivasigamani Ranjith Kumar
- Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Konarskiego 22B, 44-100 Gliwice, Poland;
| | - Angelika Banaś
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland;
| | - Katarzyna Krukiewicz
- Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Konarskiego 22B, 44-100 Gliwice, Poland;
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland;
| |
Collapse
|
40
|
Mohamed HT, El-Shinawi M, Mohamed MM. Editorial: Inflammatory tumor microenvironment: role of cytokines and virokines in breast cancer progression and metastasis. Front Cell Dev Biol 2024; 12:1414734. [PMID: 38903531 PMCID: PMC11188433 DOI: 10.3389/fcell.2024.1414734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/21/2024] [Indexed: 06/22/2024] Open
Affiliation(s)
- Hossam Taha Mohamed
- Faculty of Biotechnology, October University for Modern Sciences and Arts, Giza, Egypt
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
- Faculty of Science, Galala University, Suez, Egypt
| | | | - Mona Mostafa Mohamed
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
- Faculty of Science, Galala University, Suez, Egypt
| |
Collapse
|
41
|
Curcio C, Mucciolo G, Roux C, Brugiapaglia S, Scagliotti A, Guadagnin G, Conti L, Longo D, Grosso D, Papotti MG, Hirsch E, Cappello P, Varner JA, Novelli F. PI3Kγ inhibition combined with DNA vaccination unleashes a B-cell-dependent antitumor immunity that hampers pancreatic cancer. J Exp Clin Cancer Res 2024; 43:157. [PMID: 38824552 PMCID: PMC11143614 DOI: 10.1186/s13046-024-03080-1] [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: 03/21/2024] [Accepted: 05/24/2024] [Indexed: 06/03/2024] Open
Abstract
Phosphoinositide-3-kinase γ (PI3Kγ) plays a critical role in pancreatic ductal adenocarcinoma (PDA) by driving the recruitment of myeloid-derived suppressor cells (MDSC) into tumor tissues, leading to tumor growth and metastasis. MDSC also impair the efficacy of immunotherapy. In this study we verify the hypothesis that MDSC targeting, via PI3Kγ inhibition, synergizes with α-enolase (ENO1) DNA vaccination in counteracting tumor growth.Mice that received ENO1 vaccination followed by PI3Kγ inhibition had significantly smaller tumors compared to those treated with ENO1 alone or the control group, and correlated with i) increased circulating anti-ENO1 specific IgG and IFNγ secretion by T cells, ii) increased tumor infiltration of CD8+ T cells and M1-like macrophages, as well as up-modulation of T cell activation and M1-like related transcripts, iii) decreased infiltration of Treg FoxP3+ T cells, endothelial cells and pericytes, and down-modulation of the stromal compartment and T cell exhaustion gene transcription, iv) reduction of mature and neo-formed vessels, v) increased follicular helper T cell activation and vi) increased "antigen spreading", as many other tumor-associated antigens were recognized by IgG2c "cytotoxic" antibodies. PDA mouse models genetically devoid of PI3Kγ showed an increased survival and a pattern of transcripts in the tumor area similar to that of pharmacologically-inhibited PI3Kγ-proficient mice. Notably, tumor reduction was abrogated in ENO1 + PI3Kγ inhibition-treated mice in which B cells were depleted.These data highlight a novel role of PI3Kγ in B cell-dependent immunity, suggesting that PI3Kγ depletion strengthens the anti-tumor response elicited by the ENO1 DNA vaccine.
Collapse
Affiliation(s)
- Claudia Curcio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Piazza Nizza 44Bis, 10126, Turin, Italy
| | - Gianluca Mucciolo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Piazza Nizza 44Bis, 10126, Turin, Italy
| | - Cecilia Roux
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Piazza Nizza 44Bis, 10126, Turin, Italy
| | - Silvia Brugiapaglia
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Piazza Nizza 44Bis, 10126, Turin, Italy
| | - Alessandro Scagliotti
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Piazza Nizza 44Bis, 10126, Turin, Italy
| | - Giorgia Guadagnin
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Piazza Nizza 44Bis, 10126, Turin, Italy
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Piazza Nizza 44Bis, 10126, Turin, Italy
- Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Dario Longo
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Turin, Italy
| | - Demis Grosso
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Piazza Nizza 44Bis, 10126, Turin, Italy
| | - Mauro Giulio Papotti
- Pathology Unit, Department of Medical Sciences, University of Torino, AOU Città Della Salute E Della Scienza Di Torino, Turin, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Piazza Nizza 44Bis, 10126, Turin, Italy
- Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Paola Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Piazza Nizza 44Bis, 10126, Turin, Italy
- Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Judith A Varner
- Moores Cancer Center, Department of Pathology, University of California, San Diego, CA, USA
| | - Francesco Novelli
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Piazza Nizza 44Bis, 10126, Turin, Italy.
- Molecular Biotechnology Center, University of Torino, Turin, Italy.
| |
Collapse
|
42
|
Zhu T, Jin S, Tong D, Liu X, Liu Y, Zheng J. Enhancing the Anti-Tumor Efficacy of NK Cells on Canine Mammary Tumors through Resveratrol Activation. Animals (Basel) 2024; 14:1636. [PMID: 38891683 PMCID: PMC11171074 DOI: 10.3390/ani14111636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
In order to explore the therapeutic effect of Resveratrol (Res)-activated Natural Killer (NK) cells on canine mammary tumors, this study employed a range of assays, including wound healing, colony formation, Transwell, flow cytometry, and Western blot experiments, to investigate the impact of Res-pretreated NK cells on canine mammary tumor cells in vitro. Additionally, a tumor-bearing mouse model was utilized to further analyze the therapeutic effects of Res-pretreated NK cells in vivo. The results showed that Res enhances the capacity of NK cells to induce apoptosis, pyroptosis, and ferroptosis in canine breast tumor cells, while also augmenting their influence on the migration, invasion, and epithelial-mesenchymal transition of these cells. Furthermore, pretreatment of NK cells with Res significantly amplified their inhibitory effect on breast tumor growth in vivo and promoted tumor tissue apoptosis. Additionally, Res enhanced the recruitment of NK cells to other immune cells in the body. In summary, Res has been shown to enhance the anti-breast-tumor effect of NK cells both in vitro and in vivo, offering a new avenue for optimizing immunotherapy for canine breast tumors.
Collapse
Affiliation(s)
- Tingting Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (T.Z.); (S.J.); (D.T.); (X.L.)
| | - Shengzi Jin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (T.Z.); (S.J.); (D.T.); (X.L.)
| | - Danning Tong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (T.Z.); (S.J.); (D.T.); (X.L.)
| | - Xingyao Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (T.Z.); (S.J.); (D.T.); (X.L.)
| | - Yun Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (T.Z.); (S.J.); (D.T.); (X.L.)
| | - Jiasan Zheng
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163000, China
| |
Collapse
|
43
|
Yuan J, Lin M, Yang S, Yin H, Ouyang S, Xie H, Tang H, Ou X, Zeng Z. The therapeutic effect and targets of herba Sarcandrae on breast cancer and the construction of a prognostic signature consisting of inflammation-related genes. Heliyon 2024; 10:e31137. [PMID: 38778969 PMCID: PMC11109893 DOI: 10.1016/j.heliyon.2024.e31137] [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: 12/22/2023] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Background The prevalence of breast cancer (BRCA), which is common among women, is on the rise. This study applied network pharmacology to explore the potential mechanism of action of herba sarcandrae in BRCA and construct a prognostic signature composed of inflammation-related genes. Methods The active ingredients of herba sarcandrae were screened using the SymMap, TCMID, and TCMSP platforms, and the molecular targets were determined in the UniProt database. The "drug-active compound-potential target" network was established with Cytoscape 3.7.2. The molecular targets were subjected to disease ontology, gene ontology (GO), and Kyoto Encyclopedia of Genes (KEGG) analyses. AutoDock software was used for molecular docking. Differentially expressed genes (DEGs) related to inflammation were obtained from the BRCA Cancer Genome Atlas (TCGA) database. In the training cohort, the univariate Cox regression model was applied to preliminarily screen prognostic genes. A multigene signature was built by the least absolute shrinkage and selection operator (LASSO) regression model, followed by validation through Kaplan‒Meier, Cox, and receiver operating characteristic (ROC) analyses. Results Forty-one active compounds were identified, and 265 therapeutic targets for herba sarcandrae were predicted. GO enrichment results revealed significant enrichment of biological processes, such as response to xenobiotic stimuli, response to nutrient levels, and response to lipopolysaccharide. KEGG analysis revealed significant enrichment of pathways such as AGE-RAGE and chemical carcinogenesis receptor activation signaling pathways. In addition, the herbs Marc-Andre and rutin were shown to mediate BRCA cell proliferation and apoptosis via the interferon regulatory factor 1 (IRF1)/signal transducer and activator of transcription 3 (STAT3)/programmed death-ligand 1 (PD-L1) pathway. Sixteen inflammatory signatures, including BST2, GPR132, IL12B, IL18, IL1R1, IL2RB, IRF1, and others, were constructed, and the risk score was found to be a strong independent prognostic factor for overall survival in BRCA patients. The 16-inflammation signature was associated with several clinical features (age, clinical stage, T, and N classifications) and could reflect immune cell infiltration in tumor microenvironments with different immune cells. Conclusions Herba sarcandrae and rutin were shown to mediate BRCA cell proliferation and apoptosis via the IRF1/STAT3/PD-L1 pathway, and the 16-member inflammatory signature might be a novel biomarker for predicting BRCA patient prognosis, providing more accurate guidance for clinical treatment prognosis evaluation and having important reference value for individualized treatment selection.
Collapse
Affiliation(s)
- Jie Yuan
- Department of General Surgery, Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Minxia Lin
- Department of General Surgery, Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
| | - Shaohua Yang
- Department of General Surgery, Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Hao Yin
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Shaoyong Ouyang
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Hong Xie
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Hongmei Tang
- Pharmaceutical Department, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaowei Ou
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Zhiqiang Zeng
- Department of General Surgery, Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| |
Collapse
|
44
|
Zheng JM, Lou CX, Huang YL, Song WT, Luo YC, Mo GY, Tan LY, Chen SW, Li BJ. Associations between immune cell phenotypes and lung cancer subtypes: insights from mendelian randomization analysis. BMC Pulm Med 2024; 24:242. [PMID: 38755605 PMCID: PMC11100125 DOI: 10.1186/s12890-024-03059-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: 01/02/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024] Open
Abstract
INTRODUCTION Lung cancer is a common malignant tumor, and different types of immune cells may have different effects on the occurrence and development of lung cancer subtypes, including lung squamous cell carcinoma (LUSC) and lung adenocarcinoma (LUAD). However, the causal relationship between immune phenotype and lung cancer is still unclear. METHODS This study utilized a comprehensive dataset containing 731 immune phenotypes from the European Bioinformatics Institute (EBI) to evaluate the potential causal relationship between immune phenotypes and LUSC and LUAD using the inverse variance weighted (IVW) method in Mendelian randomization (MR). Sensitivity analyses, including MR-Egger intercept, Cochran Q test, and others, were conducted for the robustness of the results. The study results were further validated through meta-analysis using data from the Transdisciplinary Research Into Cancer of the Lung (TRICL) data. Additionally, confounding factors were excluded to ensure the robustness of the findings. RESULTS Among the final selection of 729 immune cell phenotypes, three immune phenotypes exhibited statistically significant effects with LUSC. CD28 expression on resting CD4 regulatory T cells (OR 1.0980, 95% CI: 1.0627-1.1344, p < 0.0001) and CD45RA + CD28- CD8 + T cell %T cell (OR 1.0011, 95% CI: 1.0007; 1.0015, p < 0.0001) were associated with increased susceptibility to LUSC. Conversely, CCR2 expression on monocytes (OR 0.9399, 95% CI: 0.9177-0.9625, p < 0.0001) was correlated with a decreased risk of LUSC. However, no significant causal relationships were established between any immune cell phenotypes and LUAD. CONCLUSION This study demonstrates that specific immune cell types are associated with the risk of LUSC but not with LUAD. While these findings are derived solely from European populations, they still provide clues for a deeper understanding of the immunological mechanisms underlying lung cancer and may offer new directions for future therapeutic strategies and preventive measures.
Collapse
Affiliation(s)
- Jin-Min Zheng
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi, China
| | - Chen-Xi Lou
- Department of Surgery, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Yu-Liang Huang
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi, China
| | - Wen-Tao Song
- Department of Surgery, Youjiang Medical University For Nationalities, Baise, Guangxi, China
| | - Yi-Chen Luo
- Department of thoracic surgery, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Guan-Yong Mo
- Department of thoracic surgery, Guilin Medical University, Guilin, Guangxi, China
| | - Lin-Yuan Tan
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi, China
| | - Shang-Wei Chen
- Department of thoracic surgery, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China.
| | - Bai-Jun Li
- Department of thoracic surgery, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
45
|
Yu K, Xu C, Wang F, Wang H. Identification of the new molecular subtypes related to inflammation in breast cancer. Medicine (Baltimore) 2024; 103:e38146. [PMID: 38728446 PMCID: PMC11081544 DOI: 10.1097/md.0000000000038146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
Breast cancer is a prevalent ailment among women, and the inflammatory response plays a crucial role in the management and prediction of breast cancer (BRCA). However, the new subtypes based on inflammation in BRCA research are still undefined. The databases including The Cancer Genome Atlas and gene expression omnibus were utilized to gather clinical data and somatic mutation information for approximately 1069 BRCA patients. Through Consensus Clustering, novel subtypes linked to inflammation were identified. A comparative analysis was conducted on the prognosis, and immune cell infiltration, and somatic mutation of the new subtypes. Additionally, an investigation into drug therapy and immunotherapy was conducted to distinguish high-risk individuals from low-risk ones. The findings of this investigation proposed the categorization of BRCA into innovative subtypes predicated on the inflammatory response and 6 key genes were a meaningful approach. Specifically, the low-, medium-, and high-inflammation subtypes exhibited varying degrees of association with clinicopathological features, tumor microenvironment, and prognosis. Notably, the high-inflammation subtype was characterized by a strong correlation with immunosuppressive microenvironments and a higher frequency of somatic mutations, which was an indication of poorer health. This study revealed that a brand-new classification could throw new light on the effective prognosis. The integration of multiple key genes was a new characterization that could promote more immunotherapy strategies and contribute to predicting the efficacy of the chemotherapeutic drugs.
Collapse
Affiliation(s)
- Ke Yu
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
- Department of Breast and Thyroid Surgery, Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Chi Xu
- Department of Breast and Thyroid Surgery, Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu Province, China
- Department of Gastroenterology, Affiliated of Nantong University, Nantong, Jiangsu Province, China
| | - Feng Wang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Hua Wang
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
- Department of Breast and Thyroid Surgery, Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
46
|
Guo Y, Su J, Jiang S, Xu Y, Dou B, Li T, Zhu J, He K. Transcriptomics and metabonomics study on the effect of exercise combined with curcumin supplementation on breast cancer in mice. Heliyon 2024; 10:e28807. [PMID: 38576560 PMCID: PMC10990956 DOI: 10.1016/j.heliyon.2024.e28807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/06/2024] Open
Abstract
Curcumin and exercise have been reported to show good anti-tumour effects. However, relevant research on the combined effects of physical exercise and curcumin supplementation on cancer and the underlying mechanisms is still lacking. The current study aimed to construct an anti-breast tumour mouse model using the combined effects of curcumin treatment and swimming exercise. Transcriptomic and metabolomic techniques were used to screen for differentially expressed genes and metabolites, evaluate the anticancer effects, and analyse the molecular regulatory mechanisms related to metabolism. Observation of the mouse phenotypes, including tumour appearance, in-vivo tumour imaging, and HE staining results of pathological sections, suggested a more obvious inhibitory effect of the combination of curcumin administration and exercise intervention on breast cancer than that of a single treatment. The combination treatment group had a total of 445 differentially expressed (154 upregulated and 291 downregulated) genes. Functional enrichment analysis showed the calcium signalling pathway, Wnt signalling pathway, PI3K Akt signalling pathway, and IL-17 signalling pathway to significantly participate in the anti-breast cancer process of curcumin-exercise combination treatment. Results of the intergroup differential metabolite analysis showed that the combined effect of curcumin and exercise involves two unique pathways, namely the amino sugar and nucleotide sugar metabolism, which includes chitosan, d-glucosamine 6-phosphate, l-fucose, and N-acetyl beta-mannosamine, and the amino acid biosynthesis, which includes dl-isoleucine, dl-tyrosine, and homocysteine. Collectively, the top-ranked genes and metabolites with the highest degree of associations were further revealed by O2PLS analysis. Overall, the study helped reveal the mechanism of action of curcumin-exercise combination treatment on breast cancer at multi-omics level.
Collapse
Affiliation(s)
- Yong Guo
- Post-doctoral Research Station, Harbin Sport University, Harbin, Heilongjiang, 150008, China
| | - Jinxing Su
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui, 230600, China
- Traditional Chinese Medicine Research Centre, School of Life Sciences, Anhui University, Hefei, Anhui, 230600, China
| | - Shangquan Jiang
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui, 230600, China
- Traditional Chinese Medicine Research Centre, School of Life Sciences, Anhui University, Hefei, Anhui, 230600, China
| | - Yan Xu
- School of Sports Human Science, Harbin Sport University, Harbin, Heilongjiang, 150008, China
| | - Binbin Dou
- Graduate School, Harbin Sport University, Harbin, Heilongjiang, 150008, China
| | - Ting Li
- School of Sports Human Science, Harbin Sport University, Harbin, Heilongjiang, 150008, China
| | - Jiabin Zhu
- Winter Olympics Academy, Harbin Sport University, Harbin, Heilongjiang, 150008, China
| | - Kan He
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui, 230600, China
- Traditional Chinese Medicine Research Centre, School of Life Sciences, Anhui University, Hefei, Anhui, 230600, China
| |
Collapse
|
47
|
Lu Q, Kou D, Lou S, Ashrafizadeh M, Aref AR, Canadas I, Tian Y, Niu X, Wang Y, Torabian P, Wang L, Sethi G, Tergaonkar V, Tay F, Yuan Z, Han P. Nanoparticles in tumor microenvironment remodeling and cancer immunotherapy. J Hematol Oncol 2024; 17:16. [PMID: 38566199 PMCID: PMC10986145 DOI: 10.1186/s13045-024-01535-8] [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: 12/30/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer immunotherapy and vaccine development have significantly improved the fight against cancers. Despite these advancements, challenges remain, particularly in the clinical delivery of immunomodulatory compounds. The tumor microenvironment (TME), comprising macrophages, fibroblasts, and immune cells, plays a crucial role in immune response modulation. Nanoparticles, engineered to reshape the TME, have shown promising results in enhancing immunotherapy by facilitating targeted delivery and immune modulation. These nanoparticles can suppress fibroblast activation, promote M1 macrophage polarization, aid dendritic cell maturation, and encourage T cell infiltration. Biomimetic nanoparticles further enhance immunotherapy by increasing the internalization of immunomodulatory agents in immune cells such as dendritic cells. Moreover, exosomes, whether naturally secreted by cells in the body or bioengineered, have been explored to regulate the TME and immune-related cells to affect cancer immunotherapy. Stimuli-responsive nanocarriers, activated by pH, redox, and light conditions, exhibit the potential to accelerate immunotherapy. The co-application of nanoparticles with immune checkpoint inhibitors is an emerging strategy to boost anti-tumor immunity. With their ability to induce long-term immunity, nanoarchitectures are promising structures in vaccine development. This review underscores the critical role of nanoparticles in overcoming current challenges and driving the advancement of cancer immunotherapy and TME modification.
Collapse
Affiliation(s)
- Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, China
| | - Dongquan Kou
- Department of Rehabilitation Medicine, Chongqing Public Health Medical Center, Chongqing, China
| | - Shenghan Lou
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China
| | - Amir Reza Aref
- Xsphera Biosciences, Translational Medicine Group, 6 Tide Street, Boston, MA, 02210, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Israel Canadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, USA
| | - Xiaojia Niu
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Yuzhuo Wang
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Pedram Torabian
- Cumming School of Medicine, Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Medical Sciences, University of Calgary, Calgary, AB, T2N 4Z6, Canada
| | - Lingzhi Wang
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore
| | - Gautam Sethi
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore.
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore, Republic of Singapore
| | - Franklin Tay
- The Graduate School, Augusta University, 30912, Augusta, GA, USA
| | - Zhennan Yuan
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Peng Han
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
| |
Collapse
|
48
|
Xu S, Wang Q, Ma W. Cytokines and soluble mediators as architects of tumor microenvironment reprogramming in cancer therapy. Cytokine Growth Factor Rev 2024; 76:12-21. [PMID: 38431507 DOI: 10.1016/j.cytogfr.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Navigating the intricate landscape of the tumor microenvironment (TME) unveils a pivotal arena for cancer therapeutics, where cytokines and soluble mediators emerge as double-edged swords in the fight against cancer. This review ventures beyond traditional perspectives, illuminating the nuanced interplay of these elements as both allies and adversaries in cancer dynamics. It critically evaluates the evolving paradigms of TME reprogramming, spotlighting innovative strategies that target the sophisticated network of cytokines and mediators. Special focus is placed on unveiling the therapeutic potential of novel cytokines and mediators, particularly their synergistic interactions with extracellular vesicles, which represent underexplored conduits for therapeutic targeting. Addressing a significant gap in current research, we explore the untapped potential of these biochemical players in orchestrating immune responses, tumor proliferation, and metastasis. The review advocates for a paradigm shift towards exploiting these dynamic interactions within the TME, aiming to transcend conventional treatments and pave the way for a new era of precision oncology. Through a critical synthesis of recent advancements, we highlight the imperative for innovative approaches that harness the full spectrum of cytokine and mediator activities, setting the stage for breakthrough therapies that offer heightened specificity, reduced toxicity, and improved patient outcomes.
Collapse
Affiliation(s)
- Suling Xu
- Department of Dermatology, The First Affiliated Hospital of Ningbo University School of Medicine, Ningbo, Zhejiang 315020, China.
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Wenxue Ma
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, and Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
49
|
Sang B, Fan Y, Wang X, Dong L, Gong Y, Zou W, Zhao G, He J. The prognostic value of absolute lymphocyte count and neutrophil-to-lymphocyte ratio for patients with metastatic breast cancer: a systematic review and meta-analysis. Front Oncol 2024; 14:1360975. [PMID: 38515567 PMCID: PMC10955091 DOI: 10.3389/fonc.2024.1360975] [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: 12/24/2023] [Accepted: 02/16/2024] [Indexed: 03/23/2024] Open
Abstract
Background Neutrophil-to-lymphocyte ratio (NLR) is considered a potential prognostic marker in early breast cancer. However, the prognosis of absolute lymphocyte count (ALC) and NLR in metastatic breast cancer (MBC) has been reported in a few studies, and conclusions are still conflicting. This present manuscript aims to provide further solid evidence regarding the prognostic values of ALC and NLR in MBC patients. Method Eligible studies that reported the associations between ALC or NLR and MBC were included by searching relative electronic databases. Overall survival (OS) and progression-free survival (PFS) were used as outcome measures. The hazard ratio (HR) values and 95% confidence interval (CI) of the outcome measures were collected as effect sizes, and further analysis and discussion were conducted according to the pooled HR, subgroup analysis, publication bias, and interstudy heterogeneity. Results Twenty-nine studies comprising 3,973 patients with MBC were included. According to our findings, lower ALC was significantly associated with poorer prognosis of OS (HR = 0.57, 95% CI 0.48 to 0.68) and PFS (HR = 0.68, 95% CI 0.58 to 0.79), and greater NLR was associated with poorer OS (HR = 1.50, 95% CI 1.35 to 1.67) and PFS (HR = 1.82, 95% CI 1.42 to 2.35). Furthermore, the prognostic values of ALC and NLR in MBC were also observed in the subgroup analyses regarding cutoff values and ethnicities. Conclusion Low ALC and elevated NLR were observed to be significantly associated with adverse OS and PFS in MBC, indicating that ALC and NLR may act as potential prognostic biomarkers of MBC patients. Meanwhile, our results will also provide some novel evidence and research clues for the selection and development of clinical treatment strategies for MBC patients. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42021224114.
Collapse
Affiliation(s)
- Bulin Sang
- Clinical Pharmacology Research Center, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| | - Yuxin Fan
- Clinical Pharmacology Research Center, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| | - Xurao Wang
- College of Pharmacy, Dali University, Dali, China
| | - Lixian Dong
- Clinical Pharmacology Research Center, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| | - Yuanyuan Gong
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Wenhong Zou
- Clinical Pharmacology Research Center, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| | - Guanhua Zhao
- Clinical Pharmacology Research Center, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| | - Jianchang He
- Clinical Pharmacology Research Center, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| |
Collapse
|
50
|
Gao S, Ding S, Tang Z. A preliminary mechanistic exploration of the effect of leptin on the docetaxel sensitivity of MDA‑MB‑231 triple‑negative breast cancer cells. Mol Clin Oncol 2024; 20:24. [PMID: 38410187 PMCID: PMC10895386 DOI: 10.3892/mco.2024.2722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/08/2024] [Indexed: 02/28/2024] Open
Abstract
Breast cancer is a common tumor encountered in women, and triple-negative breast cancer (TNBC) has an extremely poor prognosis. The effect of leptin on the docetaxel sensitivity of MDA-MB-231 TNBC cells has not been investigated. The present study aimed to clarify the effect of leptin and M2 tumor-associated macrophages (TAMs) on the chemosensitivity of TNBC cell lines and its possible mechanisms. In the present study, the apoptosis of the MDA-MB-231 cell line was detected at 0, 24, 48 and 72 h using a Cell Counting Kit-8 assay to determine the appropriate concentration of docetaxel as well as the IC50 value. After determining the effect of leptin on TAMs, the conditioned medium with an appropriate concentration of docetaxel was collected to treat the breast cancer cells, and flow cytometry was used to detect the cell cycle distribution and apoptosis in different treatment groups. Interleukin 8 (IL-8) expression was detected using ELISA and western blot assay. The IL-8 antibody was used to neutralize IL-8, and invasion and scratch assays were used to detect changes in invasion and migration of breast cancer cells. Statistical analysis was performed using GraphPad Prism 9.0 and SPSS 22.0. It was revealed that the apoptotic rate of MDA-MB-231 cells in the leptin-treated TAMs group was lower than that in other groups. The expression of IL-8 was notably elevated in the group treated with leptin-activated TAMs compared with that in the other groups. The neutralization of IL-8 resulted in a significant reduction in the invasive migration of MDA-MB-231 cells compared with that in the non-neutralized group.
Collapse
Affiliation(s)
- Simeng Gao
- Department of Oncology, The Central Hospital of Yongzhou, Yongzhou, Hunan 425000, P.R. China
| | - Sijuan Ding
- Department of Oncology, The Central Hospital of Yongzhou, Yongzhou, Hunan 425000, P.R. China
| | - Zhaohui Tang
- Department of Oncology, The Central Hospital of Yongzhou, Yongzhou, Hunan 425000, P.R. China
| |
Collapse
|