1
|
Mai Q, Du Q, Zeng F, Chen Y, Wang X, Zou Q, Chen Q, Huang H, Lin X, Chi C, Chen Y, Li J, Xu J, Wang S, Peng Z, Abdugheni K, Wei M, Pan C, Yao S, Liu J. Galectin-3 suppresses CD8 + T cells function via myeloid-derived suppressor cells recruitment in cervical cancer. Int J Biol Macromol 2025:143683. [PMID: 40318724 DOI: 10.1016/j.ijbiomac.2025.143683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/27/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
The tumor immune microenvironment is crucial in tumor development. Galectin-3 (GAL3), a beta-galactoside-binding lectin and tumor secretory protein, is increasingly recognized as a key mediator of immunosuppression in various cancers; however, its role in cervical cancer (CC) immune escape has not been well studied. This study investigates GAL3 regulation of myeloid-derived suppressor cells (MDSCs) through immune infiltration, flow cytometry, cytokine screening, and animal models. Particularly, GAL3 was upregulated in CC samples and was significantly correlated with lymph node metastasis, recurrence, and survival. Correlation analysis showed that GAL3 expression was correlated with the aggregation of MDSCs. Patients with high MDSCs infiltration have a poor prognosis. Mechanistically, GAL3 promoted MDSCs recruitment by activating STAT3/Akt signaling pathway. Additionally, MDSCs inhibited CD8+ T cells function by secreting interleukin-6 (IL-6) and C-X-C motif chemokine ligand 2 (CXCL2). Furthermore, GAL3 and MDSCs inhibitors diminished subcutaneous tumor in vivo by reducing MDSCs accumulation and increasing CD8+ cells infiltration. This study demonstrates that GAL3 enhances the immunosuppressive function of MDSCs in CC, leading to impaired CD8+ T cells function and poor patient prognosis. These findings identify GAL3 and MDSCs as promising targets for therapeutic intervention, providing a basis for novel immunotherapeutic approaches in CC treatment.
Collapse
Affiliation(s)
- Qiuwen Mai
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Qiqiao Du
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Feitianzhi Zeng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Yili Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Xiaojun Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Qiaojian Zou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Qianrun Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Hua Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Xiaoying Lin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Chudan Chi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Yanfei Chen
- Department of Obstetrics and Gynecology, Zhongshan Bo'ai Hospital, Zhongshan 528402, PR China
| | - Jie Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Jing Xu
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Shuyi Wang
- Department of Obstetrics and Gynecology, Qingdao Municipal Hospital, Qingdao 266011, PR China
| | - Zhangqing Peng
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China
| | - Karima Abdugheni
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Mengxun Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Chaoyun Pan
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, PR China.
| | - Shuzhong Yao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China.
| | - Junxiu Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China.
| |
Collapse
|
2
|
Diao H, Zhao F, Wu M, Zhang Y, Tao Q, Chen S, Lin D. LncRNA Expression Profiles in C6 Ceramide Treatment Reveal lnc_025370 as a Promoter in Canine Mammary Carcinoma CHMp Cells Progression. Curr Issues Mol Biol 2024; 46:14190-14203. [PMID: 39727977 DOI: 10.3390/cimb46120849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
Canine mammary carcinomas (CMCs) represent the most prevalent form of cancer in female dogs, characterized by a high incidence and mortality rate. C6 ceramide is recognized for its multifaceted anti-cancer properties, yet its specific influence on CMCs remains to be elucidated. Long noncoding RNAs (lncRNAs), now recognized as functional "dark matter" in precision oncology, are particularly intriguing, with 44% of canine lncRNAs exhibiting tissue-specific expression. In this study, we performed a thorough analysis of lncRNA expression profiles to uncover the mechanisms behind C6 ceramide's anti-cancer activity in CHMp cells. Our findings reveal that C6 ceramide notably inhibits the proliferation of CHMp cells. RNA sequencing identified 4522 lncRNAs with expression changes following C6 ceramide treatment, of which 2936 were upregulated and 1586 were downregulated. Further investigation into Lnc_025370 showed that it is predominantly nuclear-localized and is significantly downregulated by C6 ceramide treatment. Functional studies discovered that overexpression of Lnc_025370 enhances the growth and metastatic capabilities of CHMp cells, which is associated with an increase in NRG1, and concurrently diminishes the anti-cancer effectiveness of C6 ceramide in vitro. Mouse xenograft models also showed that Lnc_025370 overexpression promotes tumor growth and Ki67 expression. Together, our results suggest that Lnc_025370 acts as a pivotal target mediator of C6 ceramide's anti-cancer effects, facilitating the malignant progression of CHMp cells.
Collapse
Affiliation(s)
- Hongxiu Diao
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Fangying Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Meijin Wu
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yan Zhang
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Qianting Tao
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Shichao Chen
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Degui Lin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
3
|
Dong W, Li J, Zhuang Z. Neutrophil-related Signature Characterizes Immune Landscape and Predicts Prognosis of Invasive Breast Cancer. Biochem Genet 2024:10.1007/s10528-024-10940-0. [PMID: 39417978 DOI: 10.1007/s10528-024-10940-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 10/06/2024] [Indexed: 10/19/2024]
Abstract
As a leading prevalent malignancy, breast cancer remains a significant worldwide health issue. Recent research indicates that neutrophils play a crucial role in breast cancer development. The prognostic significance of neutrophil-related genes (NRGs) or the immune landscape of the neutrophil-related signature in invasive breast cancer (IBC) is, nevertheless, unknown. To uncover innovative therapy alternatives, the significance of the neutrophil-related signatures in IBC was evaluated here. Briefly, a prediction model based on neutrophil-related core prognostic genes and The Cancer Genome Atlas data was created (TCGA). The model may assess IBC patients' prognosis. The IBC data from the Gene Expression Omnibus (GEO) confirmed the prognostic accuracy of the model. The overall survival (OS) of patients was worse in the group with a high NRGs score compared to the group with a low NRGs score. In addition, patients with low NRGs scores were considerably more sensitive to vinorelbine, cyclophosphamide, epirubicin, gemcitabine, paclitaxel, 5-fluorouracil, docetaxel, and cisplatin. Patients with low NRGs scores responded better to CTLA-4 and PD-1 treatments. Additionally, the immune microenvironment components were more abundant in patients with low NRGs scores. Moreover, qRT-PCR results confirmed that LEF1 had a higher expression level in tumor samples compared to normal samples, whereas NRG1 and STX11 exhibited lower expression levels in tumor samples than in normal samples. These results suggest that NRGs might be utilized as biomarkers to predict the prognosis of individuals with IBC, thereby paving the way for the creation of customized therapies for IBC.
Collapse
Affiliation(s)
- Wenge Dong
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Jiejing Li
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Zhigang Zhuang
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
4
|
Hou G, Niu T, Jia A, Zhang Y, Chen X, Wei H, Jia Y, Xu Y, Li Y, Wang P, Chatterjee A. NRG1 promotes tumorigenesis and metastasis and afatinib treatment efficiency is enhanced by NRG1 inhibition in esophageal squamous cell carcinoma. Biochem Pharmacol 2023; 218:115920. [PMID: 37989416 DOI: 10.1016/j.bcp.2023.115920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a highly aggressive tumor with significant heterogeneity in incidence and outcomes. The role of Neuregulin 1 (NRG1) in ESCC and its contribution to aggressiveness remain unknown. This study aims to investigate the functions and molecular mechanisms of NRG1 in ESCC as well as the treatment strategy for ESCC with overexpression of NRG1. We firstly demonstrated the upregulation of NRG1 and a negative correlation trend between patients' overall survival (OS) and the expression level of NRG1 in esophageal cancer. And then we found NRG1 promoted cell proliferation, migration, inhibited apoptosis, and accelerated tumorigenesis and metastasis in ESCC using cell lines and xenograft models. Furthermore, we discovered that NRG1 activated the NF-κB/MMP9 signaling pathway, contributing to the metastatic phenotype in ESCC. Finally, we show that afatinib (FDA approved cancer growth blocker) could inhibit ESCC with overexpressed NRG1 and down-regulation of NRG1 along with afatinib treatment provides higher efficient strategy. This study uncovers the critical role and molecular mechanism of NRG1 in ESCC tumorigenesis and metastasis, suggesting its potential as a novel biomarker for ESCC treatment.
Collapse
Affiliation(s)
- Guiqin Hou
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| | - Tengda Niu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ang Jia
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yingying Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xunan Chen
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Huiyun Wei
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yilin Jia
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yichao Xu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yan Li
- Center of Advanced Analysis & Gene Sequencing, Zhengzhou University, Zhengzhou 450001, China
| | - Pengju Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China.
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand; School of Health Sciences and Technology, UPES, Dehradun, India.
| |
Collapse
|
5
|
Niang DGM, Gaba FM, Diouf A, Hendricks J, Diallo RN, Niang MDS, Mbengue B, Dieye A. Galectin-3 as a biomarker in breast neoplasms: Mechanisms and applications in patient care. J Leukoc Biol 2022; 112:1041-1052. [PMID: 36125083 DOI: 10.1002/jlb.5mr0822-673r] [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: 02/12/2022] [Revised: 08/26/2022] [Indexed: 12/24/2022] Open
Abstract
Galectin-3 is a member of the lectin family encoded by the LGALS3 gene on chromosome 14. It is secreted by a wide range of immune cells and mammary tumor cells. Through its activity on the tumor microenvironment, in particular on tumor-infiltrating leukocytes, galectin-3 improves the proliferation, survival, and colonizing ability of mammary neoplastic cells. Consequently, galectin-3 expression in the tumor microenvironment could worsen therapeutic outcomes of breast neoplasms and become a biomarker and a therapeutic target in combined immunotherapy in breast neoplasms. There is a limited amount of information that is available on galectin-3 in breast cancer in Africa. In this review, we analyze how galectin-3 influences the tumor microenvironment and its potential as a biomarker and therapeutic target in breast neoplasms. We aim to emphasize the significance of investigating galectin-3 in breast neoplasms in Africa based on the results of studies conducted elsewhere.
Collapse
Affiliation(s)
- Doudou Georges Massar Niang
- Division of Immunology, School of Medicine, Pharmacy and Dentistry, Cheikh Anta Diop University, Dakar, Senegal
| | - Folly Mawulolo Gaba
- Division of Immunology, School of Medicine, Pharmacy and Dentistry, Cheikh Anta Diop University, Dakar, Senegal
| | - Adame Diouf
- Division of Immunology, School of Medicine, Pharmacy and Dentistry, Cheikh Anta Diop University, Dakar, Senegal
| | - Jacobus Hendricks
- Department of Physiology and Environmental Health, University of Limpopo, Sovenga, Limpopo province, South Africa
| | - Rokhaya Ndiaye Diallo
- Division of Human Genetics, School of Medicine, Pharmacy and Dentistry, Cheikh Anta Diop University, Dakar, Senegal
| | - Maguette Deme Sylla Niang
- Division of Immunology, School of Medicine, Pharmacy and Dentistry, Cheikh Anta Diop University, Dakar, Senegal
| | - Babacar Mbengue
- Division of Immunology, School of Medicine, Pharmacy and Dentistry, Cheikh Anta Diop University, Dakar, Senegal
| | - Alioune Dieye
- Division of Immunology, School of Medicine, Pharmacy and Dentistry, Cheikh Anta Diop University, Dakar, Senegal
| |
Collapse
|
6
|
Cheng Z, Cai K, Xu C, Zhan Q, Xu X, Xu D, Zeng Q. Prognostic Value of Serum Galectin-3 in Chronic Heart Failure: A Meta-Analysis. Front Cardiovasc Med 2022; 9:783707. [PMID: 35252382 PMCID: PMC8894589 DOI: 10.3389/fcvm.2022.783707] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To evaluate the association between serum galectin-3 and all-cause death (ACD) and cardiovascular death (CVD) in patients with chronic heart failure (CHF). Methods The PubMed and Embase databases and Clinical Trials Registry (www.clinicaltrials.gov) were searched for studies with data on serum galectin-3 and ACD and CVD in CHF patients. The hazard ratios (HRs) of ACD and CVD were calculated and presented with 95% CIs. HRs were pooled using fixed effects or random effects models when appropriate. Sensitivity analysis, meta-regression and subgroup analysis were applied to find the origin of heterogeneity. Visual inspection of Begg's funnel plot and Egger's test were performed to assess the possibility publication bias. Results Pooled data included the results from 6,440 patients from 12 studies in the meta-analysis. Higher serum galectin-3 was associated with a higher risk of ACD (HR, 1.38; 95% CI, 1.14–1.67) and CVD (HR, 1.13; 95% CI, 1.02–1.25) in CHF patients. In the subgroup analyses, higher serum galectin-3 was associated with an increased risk of ACD in all subgroups. The pooled HR of the shorter follow-up group (1.78; 95% CI, 1.50–2.11) was significantly higher than the pooled HR of the longer follow-up group (1.15; 95% CI, 1.05–1.25). Sensitivity analysis of eliminating one study in each turn indicated that Koukoui et al.'s study had the largest influence on the risk of all-cause death. All-cause death publication bias was not detected (Pr>|z| = 0.35 for Begg's test and P>|t| = 0.15 for Egger's test). Conclusions Serum galectin-3 has prognostic value of both all-cause death and cardiovascular death in CHF. Serum galectin-3 could be useful for risk classification in patients with CHF. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=193399.
Collapse
Affiliation(s)
- Zhendong Cheng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Kefeng Cai
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Chaoxian Xu
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qiong Zhan
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Xingbo Xu
- Department of Cardiology and Pneumology, University Medical Center of Göttingen, Georg-August-University, Göttingen, Germany
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
- Dingli Xu
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
- *Correspondence: Qingchun Zeng
| |
Collapse
|
7
|
Abstract
Neuregulins, members of the largest subclass of growth factors of the epidermal growth factor family, mediate a myriad of cellular functions including survival, proliferation, and differentiation in normal tissues through binding to receptor tyrosine kinases of the ErbB family. However, aberrant neuregulin signaling in the tumor microenvironment is increasingly recognized as a key player in initiation and malignant progression of human cancers. In this chapter, we focus on the role of neuregulin signaling in the hallmarks of cancer, including cancer initiation and development, metastasis, as well as therapeutic resistance. Moreover, role of neuregulin signaling in the regulation of tumor microenvironment and targeting of neuregulin signaling in cancer from the therapeutic perspective are also briefly discussed.
Collapse
|
8
|
Ramadan RA, Ragab W, Assaad RS, Shaaban AE, Fayad AI. Identification of serum biomarker panel to differentiate malignant from benign thyroid nodules using multiplex bead assay. J Egypt Natl Canc Inst 2020; 32:35. [PMID: 32885338 DOI: 10.1186/s43046-020-00046-0] [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: 05/11/2020] [Accepted: 07/29/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The challenging target in the workup of thyroid nodule(s) is to exclude or diagnose thyroid cancer efficiently prior to surgical intervention. The present work studied a panel of eight serum biomarkers to differentiate benign from malignant thyroid nodules, aiming at reducing unnecessary thyroidectomy performed for inconclusive preoperative fine needle aspiration cytology. Serum interleukin-5 (IL-5), interleukin-8 (IL-8), hepatocyte growth factor (HGF), epidermal growth factor (EGF), angiopietin (Ang1), nonokine induced by interferon gamma (MIG), galectin (Gal-3), and vitamin D-binding protein (VDRP) were quantified by multiplex bead assay using Luminex xMAP technology. The study was conducted on 60 subjects of three groups (20 each; healthy controls, benign thyroid nodule, and malignant thyroid nodule). RESULTS Significant increase of the following biomarkers in the malignant group compared to the benign group was found; IL-8: 29.7 vs 8.75 pg/ml, p < 0.001, EGF: 128.7 vs 6.72 pg/ml, p < 0.001, HGF: 173.2 vs 112.2 pg/ml, p = 0.012, MIG: 776.7 vs 438 pg/ml, p = 0.023, and Ang-1: 95016 vs 33327.5 pg/ml, p = 0.014. No significant differences were detected for IL-5, Gal-3, and VDBP. Serum IL-8 and EGF showed the highest diagnostic performance individually with area under the curve (AUC) 0.849 and 0.848, respectively. The combined biomarker panels of IL-8 and EGF and IL-8, EGF, and MIG have reached a sensitivity and specificity of 95% and 65%, respectively, with a negative predictive value of 92.9%. CONCLUSIONS Serum IL-8 and EGF individually or the combined biomarker panel of IL-8, EGF, and MIG are promising tests that can help to exclude malignancy in thyroid nodule workup.
Collapse
Affiliation(s)
- Ragaa Abdelkader Ramadan
- Chemical Pathology Department, Medical Research Institute, Alexandria University, Alexandria, Egypt.
| | - Wafaa Ragab
- Chemical Pathology Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Ramy Samir Assaad
- Chemical Pathology Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Ahmed Elsayed Shaaban
- Department of Experimental and Clinical Surgery, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Amira Ibrahim Fayad
- Clinical Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
9
|
Bracun V, Aboumsallem JP, van der Meer P, de Boer RA. Cardiac Biomarkers in Patients with Cancer: Considerations, Clinical Implications, and Future Avenues. Curr Oncol Rep 2020; 22:67. [PMID: 32514994 PMCID: PMC7280346 DOI: 10.1007/s11912-020-00930-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF THE REVIEW As the number of cancer survivors increases due to early screening and modern (antineoplastic) treatments, cancer treatment associated cardiotoxicity (CTAC) is becoming an increasing health burden that affects survival and quality of life among cancer survivors. Thus, clinicians need to identify adverse events early, in an effort to take suitable measures before the occurrence of permanent or irreversible cardiac dysfunction. RECENT FINDINGS Cardiac troponin (cTn) and B-type natriuretic peptide (BNP) have been proven to detect subclinical cardiotoxicity during antineoplastic treatment. As such, these cardio-specific biomarkers could predict which patients are at risk of developing CTAC even before the start of therapy. Nevertheless, there are inconsistent data from published studies, and the recommendations regarding the use of these biomarkers and their validity are mostly based on expert consensus opinion. In this review, we summarize available literature that evaluates biomarkers of CTAC, and we encourage strategies that integrate circulating biomarkers and cardiac imaging in identifying cancer patients that are at high risk.
Collapse
Affiliation(s)
- Valentina Bracun
- Department of Cardiology, University Medical Center Groningen, University of Groningen, AB31, PO Box 30.001, 9700 RB Groningen, the Netherlands
| | - Joseph Pierre Aboumsallem
- Department of Cardiology, University Medical Center Groningen, University of Groningen, AB31, PO Box 30.001, 9700 RB Groningen, the Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, AB31, PO Box 30.001, 9700 RB Groningen, the Netherlands
| | - Rudolf A. de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, AB31, PO Box 30.001, 9700 RB Groningen, the Netherlands
| |
Collapse
|
10
|
Lin QG, Liu W, Mo YZ, Han J, Guo ZX, Zheng W, Wang JW, Zou XB, Li AH, Han F. Development of prognostic index based on autophagy-related genes analysis in breast cancer. Aging (Albany NY) 2020; 12:1366-1376. [PMID: 31967976 PMCID: PMC7053636 DOI: 10.18632/aging.102687] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/25/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Autophagy is a self-digesting process that can satisfy the metabolic needs of cells, and is closely related to development of cancer. However, the effect of autophagy-related genes (ARGs) on the prognosis of breast cancer remains unclear. RESULTS We first found that 27 ARGs were significantly associated with overall survival in breast cancer. The prognosis-related ARGs signature established using the Cox regression model consists of 12 ARGs that can be divided patients into high-risk and low-risk groups. The overall survival of patients with high-risk scores (HR 3.652, 2.410-5.533; P < 0.001) was shorter than patients with low-risk scores. The area under the receiver operating characteristic (ROC) curve for 1-year, 3-year, and 5-year survival rates were 0.739, 0.727, and 0.742, respectively. CONCLUSION The12-ARGs marker can predict the prognosis of breast cancer and thus help individualized treatment of patients at different risks. METHODS Based on the TCGA dataset, we integrated the expression profiles of ARGs in 1,039 breast cancer patients. Differentially expressed ARGs and survival-related ARGs were evaluated by computational difference algorithm and COX regression analysis. In addition, we also explored the mutations in these ARGs. A new prognostic indicator based on ARGs was developed using multivariate COX analysis.
Collapse
Affiliation(s)
- Qing-Guang Lin
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Wei Liu
- Department of Breast, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou 510220, Guangdong, China
| | - Yu-Zhen Mo
- Department of Radiotherapy, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou 510220, Guangdong, China
| | - Jing Han
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Zhi-Xing Guo
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Wei Zheng
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Jian-Wei Wang
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Xue-Bin Zou
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - An-Hua Li
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Feng Han
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, China
| |
Collapse
|
11
|
Huang W, Wei X, Wei Y, Feng R. Biology of Tumor Associated Macrophages in Diffuse Large B Cell Lymphoma. DNA Cell Biol 2018; 37:947-952. [PMID: 30403536 DOI: 10.1089/dna.2018.4374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The tumor associated microenvironment is known to play a vital role during the development and progression of different malignant tumors. As a part of tumor microenvironment, tumor associated macrophages (TAMs) are crucial for the genesis, proliferation, metastasis, and survival of tumor cells. Recently, more and more studies showed that TAMs were related with poor clinical status and survival in patients with diffuse large B cell lymphoma (DLBCL). Considering the complex roles which TAMs play in the tumor microenvironment of DLBCL, the aim of this study was to review the biological mechanisms between TAMs and DLBCL cells, including extracellular matrix remodeling and angiogenesis promotion, tumor promotion, immune suppression, and phagocytosis inhibition. This review will help us to further understand the comprehensive impact of TAMs on DLBCL and explore possible prognostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Weimin Huang
- Department of Hematology, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Xiaolei Wei
- Department of Hematology, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Yongqiang Wei
- Department of Hematology, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Ru Feng
- Department of Hematology, Nanfang Hospital, Southern Medical University , Guangzhou, China
| |
Collapse
|
12
|
Piek A, Du W, de Boer RA, Silljé HHW. Novel heart failure biomarkers: why do we fail to exploit their potential? Crit Rev Clin Lab Sci 2018; 55:246-263. [PMID: 29663841 DOI: 10.1080/10408363.2018.1460576] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Plasma biomarkers are useful tools in the diagnosis and prognosis of heart failure (HF). In the last decade, numerous studies have aimed to identify novel HF biomarkers that would provide superior and/or additional diagnostic, prognostic, or stratification utility. Although numerous biomarkers have been identified, their implementation in clinical practice has so far remained largely unsuccessful. Whereas cardiac-specific biomarkers, including natriuretic peptides (ANP and BNP) and high sensitivity troponins (hsTn), are widely used in clinical practice, other biomarkers have not yet proven their utility. Galectin-3 (Gal-3) and soluble suppression of tumorigenicity 2 (sST2) are the only novel HF biomarkers that are included in the ACC/AHA HF guidelines, but their clinical utility still needs to be demonstrated. In this review, we will describe natriuretic peptides, hsTn, and novel HF biomarkers, including Gal-3, sST2, human epididymis protein 4 (HE4), insulin-like growth factor-binding protein 7 (IGFBP-7), heart fatty acid-binding protein (H-FABP), soluble CD146 (sCD146), interleukin-6 (IL-6), growth differentiation factor 15 (GDF-15), procalcitonin (PCT), adrenomedullin (ADM), microRNAs (miRNAs), and metabolites like 5-oxoproline. We will discuss the biology of these HF biomarkers and conclude that most of them are markers of general pathological processes like fibrosis, cell death, and inflammation, and are not cardiac- or HF-specific. These characteristics explain to a large degree why it has been difficult to relate these biomarkers to a single disease. We propose that, in addition to clinical investigations, it will be pivotal to perform comprehensive preclinical biomarker investigations in animal models of HF in order to fully reveal the potential of these novel HF biomarkers.
Collapse
Affiliation(s)
- Arnold Piek
- a Department of Cardiology , University Medical Center Groningen, University of Groningen , Groningen , The Netherlands
| | - Weijie Du
- a Department of Cardiology , University Medical Center Groningen, University of Groningen , Groningen , The Netherlands.,b Department of Pharmacology, College of Pharmacy , Harbin Medical University , Harbin , China
| | - Rudolf A de Boer
- a Department of Cardiology , University Medical Center Groningen, University of Groningen , Groningen , The Netherlands
| | - Herman H W Silljé
- a Department of Cardiology , University Medical Center Groningen, University of Groningen , Groningen , The Netherlands
| |
Collapse
|
13
|
Yamashita N, Saito N, Zhao S, Terai K, Hiruta N, Park Y, Bujo H, Nemoto K, Kanno Y. Heregulin-induced cell migration is promoted by aryl hydrocarbon receptor in HER2-overexpressing breast cancer cells. Exp Cell Res 2018; 366:34-40. [PMID: 29501698 DOI: 10.1016/j.yexcr.2018.02.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 02/16/2018] [Accepted: 02/26/2018] [Indexed: 12/19/2022]
Abstract
HER2 overexpression accounts for approximately 15-20% of all breast cancers. We have shown that HER2 overexpression leads to elevated expression of the aryl hydrocarbon receptor (AhR) in breast cancer cells. In this study, firstly, we showed that AhR expression was up-regulated by treatment with the HER3 ligand heregulin (HRG) in HER2-overexpressing breast cancer cell lines. Induction of AhR was mediated by transcriptional activation of the region of AhR promoter corresponding to - 190 to - 100 bp. In addition, HRG treatment elicited nuclear translocation of AhR. To investigate the role of AhR in HRG-HER2/HER3 signaling in HER2-overexpressing cells, we established AhR knockout (KO) HER2-overexpressing cells to perform wound-healing assays. HRG-induced cell migration was markedly attenuated by AhR KO. HRG-induced cell migration was associated with increased expression of the inflammatory cytokines interleukin (IL)-6 and IL-8 in wild type cells, but not in AhR KO cells. These results elucidate that AhR is an important factor for the malignancy in HER2 overexpressing breast cancers.
Collapse
Affiliation(s)
- Naoya Yamashita
- Department of Molecular Toxicology, Faculty of Pharmaceutical Sciences, Toho University, Miyama 2-2-1, Funabashi, Chiba 274-8510, Japan
| | - Nao Saito
- Department of Molecular Toxicology, Faculty of Pharmaceutical Sciences, Toho University, Miyama 2-2-1, Funabashi, Chiba 274-8510, Japan
| | - Shuai Zhao
- Department of Molecular Toxicology, Faculty of Pharmaceutical Sciences, Toho University, Miyama 2-2-1, Funabashi, Chiba 274-8510, Japan
| | - Kensuke Terai
- Department of Clinical-Laboratory and Experimental-Research Medicine, Toho University Sakura Medical Center, Shimoshizu 564-1, Sakura, Chiba 285-8741, Japan; Department of Surgical Pathology, Toho University Sakura Medical Center, Shimoshizu 564-1, Sakura, Chiba 285-8741, Japan
| | - Nobuyuki Hiruta
- Department of Clinical-Laboratory and Experimental-Research Medicine, Toho University Sakura Medical Center, Shimoshizu 564-1, Sakura, Chiba 285-8741, Japan; Department of Surgical Pathology, Toho University Sakura Medical Center, Shimoshizu 564-1, Sakura, Chiba 285-8741, Japan
| | - Youngjin Park
- Department of Surgery, Toho University Sakura Medical Center, Shimoshizu 564-1, Sakura, Chiba 285-8741, Japan; Department of Breast and Endocrine Surgery, Tohoku Medical and Pharmaceutical University, Fukumuro 1-15-1, Miyagino-ku, Sendai, Miyagi prefecture, 983-8536, Japan
| | - Hideaki Bujo
- Department of Clinical-Laboratory and Experimental-Research Medicine, Toho University Sakura Medical Center, Shimoshizu 564-1, Sakura, Chiba 285-8741, Japan
| | - Kiyomitsu Nemoto
- Department of Molecular Toxicology, Faculty of Pharmaceutical Sciences, Toho University, Miyama 2-2-1, Funabashi, Chiba 274-8510, Japan
| | - Yuichiro Kanno
- Department of Breast and Endocrine Surgery, Tohoku Medical and Pharmaceutical University, Fukumuro 1-15-1, Miyagino-ku, Sendai, Miyagi prefecture, 983-8536, Japan.
| |
Collapse
|