1
|
Jin Y, Xie X, Li H, Zhang M. The role of homeobox gene Six1 in cancer progression and its potential as a therapeutic target: A review. Int J Biol Macromol 2025; 308:142666. [PMID: 40164243 DOI: 10.1016/j.ijbiomac.2025.142666] [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: 11/04/2024] [Revised: 03/19/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
The sine oculis homeobox gene 1 (Six1), a member of the Six transcription factor family, specifically binds to defined DNA regions, regulates target gene expression, and plays a crucial role in various tissue and organ development processes. Moreover, Six1 is a critical factor in cancer progression and prognosis making it a central focus in cancer research. Consequently, a comprehensive review of involvement of the Six1 gene in cancer research has a high relevance. This review synthesizes findings from other researches, examines the gene structure and protein functionality of Six1, summarizes its relationship with various cancers, elucidates its mechanisms in promoting tumor progression and development, explores potential possibilities for targeting Six1 as a therapeutic approach for cancer treatment. Six1 is correlated with tumor malignancy and poor prognosis, plays a critical role in promoting tumor cell proliferation, invasion, metastasis, and energy metabolism. Targeting Six1 degradation or expression can potentially suppress tumor progression. This review aims to enhance our understanding of the function and significance of Six1 in cancers while providing a valuable reference for Six1-based cancer diagnosis, prognosis, and therapeutic interventions. This knowledge will facilitate more in-depth oncology research related to Six1, particularly in identifying drug resistance mechanisms and developing precision-targeted therapies.
Collapse
Affiliation(s)
- Yong Jin
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China; Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Xinran Xie
- School of Basic Medicine sciences, Inner Mongolia Medical University, Hohhot, China
| | - Hongbin Li
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China; Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China.
| | - Manling Zhang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China; Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China.
| |
Collapse
|
2
|
Gramantieri L, Montagner A, Arleo A, Suzzi F, Bassi C, Tovoli F, Bruccoleri M, Alimenti E, Fornari F, Iavarone M, Negrini M, Piscaglia F, Giovannini C. Early CTLA4 increase in CD45+ blood cells: an emerging biomarker of atezolizumab-bevacizumab resistance and worse survival in advanced hepatocarcinoma. ESMO Open 2025; 10:104289. [PMID: 40048814 PMCID: PMC11928801 DOI: 10.1016/j.esmoop.2025.104289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Advanced hepatocellular carcinoma (HCC) has a dismal prognosis; however, the introduction of atezolizumab-bevacizumab combination has improved overall survival and novel immune checkpoint inhibitors are entering the clinics. Despite more therapeutic options being available, no biomarker guides treatment choice. Indeed, tissue-based analyses and complex analytical procedures hinder clinical translation. We explored the informativeness of a simple, non-invasive, repeatable cytofluorimetric assay on peripheral blood to predict response and survival in HCC patients treated with atezolizumab-bevacizumab. MATERIALS AND METHODS Twenty-five cirrhotic patients, 50 HCC patients undergoing atezolizumab-bevacizumab and an independent validation cohort of 25 HCC patients were subjected to a cytofluorimetric study of peripheral white blood cells (WBCs) to assess baseline programmed death-ligand 1-positive (PD-L1+) and cytotoxic T-lymphocyte antigen 4-positive (CTLA4+) cell percentage in the different populations and their early on-treatment variations. Immunophenotypes were evaluated against treatment response. RNA sequencing followed by RT-PCR validation were used to elucidate the molecular correlates of immunophenotypic observations. RESULTS PD-L1+ cell percentage did not predict response either at baseline or when evaluating treatment-induced early changes. Conversely, the percentage of CTLA4+ lymphocytes at baseline showed a predictive significance (35.37 in responders versus 31.5 in non-responders, P = 0.03). More interestingly, the early CTLA4+ changes during treatment in lymphocytes (responders 0.95 versus non-responders 1.08, P = 0.05), monocytes (responders 0.95 versus non-responders 1.04, P = 0.03), granulocytes (responders 0.94 versus non-responders 1.14, P = 0.001) and, even stronger, the early CTLA4+ percentage change in the whole WBCs displayed a predictive significance in terms of time to progression (TTP) (P < 0.0001) and overall survival (OS) (P = 0.005). The immunophenotypic findings correlated with transcriptional modulation of CTLA4 target genes and genes involved in immune response. CONCLUSIONS A repeatable, easy, non-invasive blood test predicts response to immunotherapy in patients with HCC, both in terms of TTP and OS. CTLA4+ cell percentage increase in non-responders suggests a possible resistance mechanism which deserves attention as a druggable target.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/mortality
- Carcinoma, Hepatocellular/blood
- Carcinoma, Hepatocellular/pathology
- Liver Neoplasms/drug therapy
- Liver Neoplasms/mortality
- Liver Neoplasms/blood
- Liver Neoplasms/pathology
- Male
- CTLA-4 Antigen/blood
- CTLA-4 Antigen/metabolism
- Female
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Middle Aged
- Bevacizumab/therapeutic use
- Bevacizumab/pharmacology
- Biomarkers, Tumor/blood
- Aged
- Drug Resistance, Neoplasm
- Leukocyte Common Antigens/metabolism
- Leukocyte Common Antigens/blood
Collapse
Affiliation(s)
- L Gramantieri
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - A Montagner
- Department of Medical and Surgical Sciences, Bologna University, Bologna, Italy
| | - A Arleo
- Department of Medical and Surgical Sciences, Bologna University, Bologna, Italy
| | - F Suzzi
- Department of Medical and Surgical Sciences, Bologna University, Bologna, Italy
| | - C Bassi
- Department of Translational Medicine and Laboratorio per le Tecnologie delle Terapie Avanzate (LTTA) Centre, University of Ferrara, Ferrara, Italy
| | - F Tovoli
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences, Bologna University, Bologna, Italy
| | - M Bruccoleri
- Division of Gastroenterology and Hepatology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - E Alimenti
- Division of Gastroenterology and Hepatology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - F Fornari
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - M Iavarone
- Division of Gastroenterology and Hepatology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - M Negrini
- Department of Translational Medicine and Laboratorio per le Tecnologie delle Terapie Avanzate (LTTA) Centre, University of Ferrara, Ferrara, Italy
| | - F Piscaglia
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences, Bologna University, Bologna, Italy
| | - C Giovannini
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences, Bologna University, Bologna, Italy.
| |
Collapse
|
3
|
Lu Z, Dong H, Tu Z, Liu H. Expression, molecular mechanisms and therapeutic potentials of ATF1 in cancers. Life Sci 2025; 360:123256. [PMID: 39580140 DOI: 10.1016/j.lfs.2024.123256] [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: 09/17/2024] [Revised: 11/06/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024]
Abstract
Activating transcription factor 1 (ATF1) is a crucial cellular regulator, with its misregulation implicated in numerous cancers. As a key player in the ATF/CREB family, ATF1 modulates gene expression in response to extracellular signals, significantly impacting cancer progression. This review examines ATF1's structural features, its role in tumorigenesis, and its potential therapeutic applications. Data from various databases consistently show ATF1 overexpression in diverse cancers, associated with poor prognosis and aggressive phenotypes. The review explores ATF1's complex regulatory mechanisms, influencing cell proliferation, apoptosis, migration, invasion, and therapeutic resistance, and its interactions with regulatory networks. Emerging strategies targeting ATF1, such as engineered antibodies, natural compounds, and small molecule inhibitors, show efficacy in preclinical models. ATF1 may also act as a biomarker for personalized therapeutic response and resistance. Future research should focus on ATF1's role in the tumor microenvironment and its interaction with the immune system, potentially leading to new immunotherapeutic strategies. A deeper understanding of ATF1 could enhance cancer treatment and patient outcomes.
Collapse
Affiliation(s)
- Ziwen Lu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Hangyu Dong
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Zhigang Tu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| | - Hanqing Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
4
|
Rahman MO, Das A, Naeem N, Jabeen-E-Tahnim, Hossain MA, Alam MN, Azad AKM, Alyami SA, Alotaibi N, Al-Moisheer AS, Moni MA. An Integrated Framework to Identify Prognostic Biomarkers and Novel Therapeutic Targets in Hepatocellular Carcinoma-Based Disabilities. BIOLOGY 2024; 13:966. [PMID: 39765633 PMCID: PMC11673266 DOI: 10.3390/biology13120966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/11/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025]
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent malignant tumors globally, significantly affecting liver functions, thus necessitating the identification of biomarkers and effective therapeutics to improve HCC-based disabilities. This study aimed to identify prognostic biomarkers, signaling cascades, and candidate drugs for the treatment of HCC through integrated bioinformatics approaches such as functional enrichment analysis, survival analysis, molecular docking, and simulation. Differential expression and functional enrichment analyses revealed 176 common differentially expressed genes from two microarray datasets, GSE29721 and GSE49515, significantly involved in HCC development and progression. Topological analyses revealed 12 hub genes exhibiting elevated expression in patients with higher tumor stages and grades. Survival analyses indicated that 11 hub genes (CCNB1, AURKA, RACGAP1, CEP55, SMC4, RRM2, PRC1, CKAP2, SMC2, UHRF1, and FANCI) and three transcription factors (E2F1, CREB1, and NFYA) are strongly linked to poor patient survival. Finally, molecular docking and simulation identified seven candidate drugs with stable complexes to their target proteins: tozasertib (-9.8 kcal/mol), tamatinib (-9.6 kcal/mol), ilorasertib (-9.5 kcal/mol), hesperidin (-9.5 kcal/mol), PF-562271 (-9.3 kcal/mol), coumestrol (-8.4 kcal/mol), and clofarabine (-7.7 kcal/mol). These findings suggest that the identified hub genes and TFs could serve as valuable prognostic biomarkers and therapeutic targets for HCC-based disabilities.
Collapse
Affiliation(s)
- Md. Okibur Rahman
- Department of Pharmacy, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Asim Das
- Department of Pharmacy, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Nazratun Naeem
- Department of Pharmacy, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Jabeen-E-Tahnim
- Department of Pharmacy, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Md. Ali Hossain
- Department of Computer Science & Engineering, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
- Department of Computer Science & Engineering, Daffodil International University, Dhaka 1216, Bangladesh
| | - Md. Nur Alam
- Department of Pharmacy, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - AKM Azad
- Department of Mathematics & Statistics, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13318, Saudi Arabia
| | - Salem A. Alyami
- Department of Mathematics & Statistics, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13318, Saudi Arabia
| | - Naif Alotaibi
- Department of Mathematics & Statistics, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13318, Saudi Arabia
| | - A. S. Al-Moisheer
- Department of Mathematics & Statistics, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13318, Saudi Arabia
| | - Mohammod Ali Moni
- Artificial Intelligence and Cyber Futures Institute, Charles Sturt University, Bathurst, NSW 2795, Australia
| |
Collapse
|
5
|
Chen J, Huang X, Zhang S, Zhu X. ATF4 inhibits tumor development and mediates p-GCN2/ASNS upregulation in colon cancer. Sci Rep 2024; 14:13042. [PMID: 38844625 PMCID: PMC11156644 DOI: 10.1038/s41598-024-63895-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024] Open
Abstract
Colon cancer (CC) is a highly malignant tumor with a high incidence and poor prognosis. This study aimed to explore the function and molecular mechanisms of activating transcription factor 4 (ATF4) in CC. The expression levels of ATF4, GCN2, and ASNS in CC tissues were measured using immunohistochemistry (IHC) and reverse transcription quantitative PCR (RT-qPCR). Cell counting kit-8 (CCK-8), clone formation, transwell, and flow cytometry assays were conducted to assess cell viability, clonogenicity, migration, invasion, cell cycle, and apoptosis, respectively, in the ATF4 knockdown and overexpression SW480 cell lines. The effect of ATF4 on the expression of GCN2 and ASNS was detected using RT-qPCR, Chip-qPCR, and western blotting. ATF4, GCN2, and ASNS were expressed at low levels in CC tissues, and all had a significant negative correlation with tumor diameter. ATF4 knockdown promoted cell proliferation, invasion, and S-phase cell cycle and inhibited apoptosis in SW480 cells. In contrast, ATF4 overexpression had the opposite effect. Furthermore, ATF4 overexpression enhanced ATF4 binding to the ASNS promoter region. ATF4 knockdown significantly inhibited the expression of p-GCN2 and ASNS, whereas ATF4 overexpression significantly upregulated their expression. ATF4 inhibited CC cell viability, clone formation ability, migration, and invasion and promoted apoptosis, possibly by regulating the expression of p-GCN2 and ASNS. Our study provides a novel potential therapeutic target for the treatment of CC.
Collapse
Affiliation(s)
- Jiawei Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Qingxiu District, Nanning, 530021, Guangxi, China
- Department of Radiation Oncology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Xiaopeng Huang
- Department of Radiation Oncology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Shuai Zhang
- Department of Radiation Oncology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China.
| | - Xiaodong Zhu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Qingxiu District, Nanning, 530021, Guangxi, China.
- Department of Oncology, Wuming Hospital of Guangxi Medical University, Nanning, Guangxi, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, China.
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, Guangxi, China.
| |
Collapse
|
6
|
Zhan S, Qiu M, Wei X, Wei J, Qin L, Jiang B, Wen Q, Chen P, Lin Q, Wei X, Zhou Z, Jiang Y, Liang X, Li R, Liu Y, Yu H. Potentially functional genetic variants in ferroptosis-related CREB3 and GALNT14 genes predict survival of hepatitis B virus-related hepatocellular carcinoma. Cancer Med 2024; 13:e6848. [PMID: 38151984 PMCID: PMC10807646 DOI: 10.1002/cam4.6848] [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: 09/09/2023] [Revised: 11/19/2023] [Accepted: 11/27/2023] [Indexed: 12/29/2023] Open
Abstract
BACKGROUND Ferroptosis is a known crucial player in the development of cancers. However, the effect of single nucleotide polymorphisms (SNPs) in ferroptosis-related genes on survival in hepatitis B virus (HBV)-related hepatocellular carcinoma (HBV-HCC) patients remains unknown. METHODS We used two-stage multivariable Cox proportional hazards regression analyses to estimate the associations between 48,774 SNPs in 480 ferroptosis-related genes and overall survival (OS) of 866 HBV-HCC patients. RESULTS We identified that two potentially functional SNPs (CREB3 rs10814274 C > T and GALNT14 rs17010547 T > C) were significantly independently associated with the OS of HBV-HCC patients (CT + TT verse CC, hazards ratio (HR) = 0.77, 95% confidence interval (CI) = 0.67-0.89, p < 0.001 for rs10814274 and TC + CC verse TT, HR = 0.66, 95% CI = 0.53-0.82, p < 0.001 for rs17010547, respectively). Additional joint assessment of protective genotypes of these two SNPs showed that patients with 1-2 protective genotypes had a significantly better OS compared with those carrying 0 protective genotypes (HR = 0.56, 95% CI = 0.45-0.70, p < 0.001). Moreover, the expression quantitative trait loci (eQTL) analysis revealed that the survival-associated SNP rs10814274 T allele was significantly correlated with reduced CREB3 transcript levels in both normal liver tissues and whole blood cells, while the GALNT14 rs17010547 C allele had a significant correlation with increased GALNT14 transcript levels in whole blood cells. CONCLUSION These results suggest that genetic variants of CREB3 and GALNT14 may affect the survival of HBV-HCC patients, likely via transcriptional regulation of respective genes. However, further studies are required to confirm these findings.
Collapse
Affiliation(s)
- Shicheng Zhan
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningChina
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
| | - Moqin Qiu
- Department of Respiratory OncologyGuangxi Medical University Cancer HospitalNanningChina
| | - Xueyan Wei
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningChina
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
| | - Junjie Wei
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningChina
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
| | - Liming Qin
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningChina
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
| | - Binbin Jiang
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningChina
| | - Qiuping Wen
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningChina
| | - Peiqin Chen
- Editorial Department of Chinese Journal of Oncology Prevention and TreatmentGuangxi Medical University Cancer HospitalNanningChina
| | - Qiuling Lin
- Department of Clinical ResearchGuangxi Medical University Cancer HospitalNanningChina
| | - Xiaoxia Wei
- Department of Clinical ResearchGuangxi Medical University Cancer HospitalNanningChina
| | - Zihan Zhou
- Department of Cancer Prevention and ControlGuangxi Medical University Cancer HospitalNanningChina
| | - Yanji Jiang
- Scientific Research DepartmentGuangxi Medical University Cancer HospitalNanningChina
| | - Xiumei Liang
- Department of Disease Process ManagementGuangxi Medical University Cancer HospitalNanningChina
| | - Runwei Li
- Department of Civil Engineering, College of EngineeringNew Mexico State UniversityLas CrucesNew MexicoUSA
| | - Yingchun Liu
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningChina
- Key Cultivated Laboratory of Cancer Molecular Medicine of Guangxi Health CommissionGuangxi Medical University Cancer HospitalNanningChina
| | - Hongping Yu
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningChina
- Key Cultivated Laboratory of Cancer Molecular Medicine of Guangxi Health CommissionGuangxi Medical University Cancer HospitalNanningChina
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University)Ministry of EducationNanningChina
| |
Collapse
|