1
|
Jiang L, Shen M, Zhang S, Zhang J, Shi Y, Gu Y, Yang T, Fu Q, Wang B, Chen Y, Xu K, Chen H. A regulatory variant rs9379874 in T1D risk region 6p22.2 affects BTN3A1 expression regulating T cell function. Acta Diabetol 2025; 62:695-706. [PMID: 39417845 DOI: 10.1007/s00592-024-02389-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE Genome-wide association studies (GWAS) have identified that 6p22.2 region is associated with type 1 diabetes (T1D) risk in the Chinese Han population. This study aims to reveal associations between this risk region and T1D subgroups and related clinical features, and further identify causal variant(s) and target gene(s) in this region. METHODS 2608 T1D and 4814 healthy controls were recruited from East, Central, and South China. Baseline data and genotyping for rs4320356 were collected. The most likely causal variant and gene were identified by bioinformatics analysis, dual-luciferase reporter assays, expression quantitative trait loci (eQTL), and functional annotation of the non-coding region within the 6p22.2 region. RESULTS The leading variant rs4320356 in the 6p22.2 region was associated with T1D risk in the Chinese and Europeans. However, this variant was not significantly associated with islet function or autoimmunity. In silico analysis suggested rs9379874 was the most potential causal variant for T1D risk among thymus, spleen, and T cells, overlapping with the enhancer-related histone mark in multiple T cell subsets. Dual luciferase reporter assay and eQTL showed that the T allele of rs9379874 increased BTN3A1 expression by binding to FOXA1. Public single-cell RNA sequencing analysis indicated that BTN3A1 was related to T-cell activation, ATP metabolism, and cytokine metabolism pathways, which might contribute to T1D development. CONCLUSION This study indicates that a functional variant rs9379874 regulates BTN3A1 expression, expanding the genomic landscape of T1D risk and offering a potential target for developing novel therapies.
Collapse
Affiliation(s)
- Liying Jiang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Department of Rehabilitation Medicine, Lishui People's Hospital, Lishui, 323000, Zhejiang, China
| | - Min Shen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Saisai Zhang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jie Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yun Shi
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yong Gu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Tao Yang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Qi Fu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Bingwei Wang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yang Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Kuanfeng Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Heng Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
2
|
Yang L, Ding C, Tuo M, Chu T, Liu P. METTL3 enhances esophageal squamous cell carcinoma progression by suppressing ferroptosis through the PBX3/CA9 cascade. Pathol Res Pract 2025; 269:155865. [PMID: 40048803 DOI: 10.1016/j.prp.2025.155865] [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: 08/15/2024] [Revised: 01/21/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND N6-methyladenosine (m6A) modification controls various processes during tumorigenesis. Although METTL3 functions as a pro-tumorigenic driver in esophageal squamous cell carcinoma (ESCC), its mechanisms are largely unknown. METHODS mRNA expression was detected by quantitative PCR, and protein expression was assessed by immunoblotting. Cell motility, invasiveness, and apoptosis were analyzed by wound-healing assay, transwell assay and flow cytometry, respectively. Cell ferroptosis was assessed by detecting the contents of ROS, MDA and Fe2+. The METTL3/PBX3 and PBX3/CA9 relationships were validated by luciferase, MeRIP or ChIP assay. The effect of METTL3 on tumor growth was tested by xenograft studies. RESULTS METTL3 was enhanced in ESCC tumors and cells, and its deficiency suppressed ESCC cell migration and invasion and promoted cell apoptosis and ferroptosis. Additionally, METTL3 deficiency caused growth inhibition of ESCC xenografts in vivo. METTL3 enhanced m6A modification of PBX3 mRNA. PBX3 was identified as a mediator of METTL3 function in modulating ESCC cell phenotypes. PBX3 promoted CA9 transcription, and METTL3 positively regulated CA9 through PBX3. PBX3 deficiency impeded ESCC cell migration and invasion and enhanced cell apoptosis and ferroptosis by downregulating CA9. CONCLUSION Our study elucidates a novel mechanism, the METTL3/PBX3/CA9 cascade, underlying the oncogenic activity of METTL3 in ESCC. The novel cascade may represent the potential target for ESCC therapy in the future.
Collapse
Affiliation(s)
- Lingxia Yang
- Department of Gastroenterology, Jingmen Traditional Chinese Medicine Hospital, Jingmen 448000, China.
| | - Chang'e Ding
- Department of Blood Transfusion, Jingmen Traditional Chinese Medicine Hospital, Jingmen 448000, China
| | - Mengjie Tuo
- Department of Pediatrics, Jingmen Traditional Chinese Medicine Hospital, Jingmen 448000, China
| | - Tiandong Chu
- Department of Surgery, Jingmen Traditional Chinese Medicine Hospital, Jingmen 448000, China
| | - Ping Liu
- Department of Gastroenterology, Jingmen Traditional Chinese Medicine Hospital, Jingmen 448000, China
| |
Collapse
|
3
|
Xiao ZW, Zeng YC, Ji LT, Yuan JT, Li L. Nitric oxide synthase 1 inhibits the progression of esophageal cancer through interacting with nitric oxide synthase 1 adaptor protein. World J Gastrointest Oncol 2025; 17:103843. [PMID: 40235872 PMCID: PMC11995332 DOI: 10.4251/wjgo.v17.i4.103843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/22/2025] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Esophageal cancer (ESCA) is among the most prevalent and lethal tumors globally. While nitric oxide synthase 1 (NOS1) is recognized for its important involvement in various cancers, its specific function in ESCA remains unclear. AIM To explore the potential role and underlying mechanisms of NOS1 in ESCA. METHODS Survival rates were analyzed using GeneCards and Gene Expression Profiling Interactive Analysis. The effects and mechanisms of NOS1 on ESCA cells were evaluated via the Cell Counting Kit-8 assay, scratch assay, Transwell assay, flow cytometry, quantitative polymerase chain reaction, western blotting, and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick end labeling staining. The protein interaction network was used to screen the interacting proteins of NOS1 and validate these interactions through co-immunoprecipitation and dual luciferase assays. Additionally, a nude mouse xenograft model was established to evaluate the effect of NOS1 in vivo. RESULTS The survival rate of patients with ESCA with high NOS1 expression was higher than that of patients with low NOS1 expression. NOS1 expression in ESCA cell lines was lower than that in normal esophageal epithelial cells. Overexpression of NOS1 (oe-NOS1) inhibited proliferation, invasion, and migration abilities in ESCA cell lines, resulting in decreased autophagy levels and increased apoptosis, pyroptosis, and ferroptosis. Protein interaction studies confirmed the interaction between NOS1 and NOS1 adaptor protein (NOS1AP). Following oe-NOS1 and the silencing of NOS1AP, levels of P62 and microtubule-associated protein 1 light chain 3 beta increased both in vitro and in vivo. Furthermore, the expression levels of E-cadherin, along with the activation of phosphatidylinositol 3-kinase (PI3K) and protein kinase B (AKT), were inhibited in ESCA cell lines. CONCLUSION NOS1 and NOS1 proteins interact to suppress autophagy, activate the PI3K/AKT pathway, and exert anti-cancer effects in ESCA.
Collapse
Affiliation(s)
- Zi-Wei Xiao
- College of Medical, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Ying-Chao Zeng
- College of Medical, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Lin-Tao Ji
- College of Medical, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Jia-Tao Yuan
- College of Medical, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Lin Li
- College of Medical, Hunan Normal University, Changsha 410081, Hunan Province, China
| |
Collapse
|
4
|
Duan Q, Wang W, Xiong H, Xiao J, Xiao H, Zhu F, Lu H. JAK2/ULK1 axis promotes cervical cancer progression by autophagy induction and SRPK1 phosphorylation. Oncogene 2025; 44:587-600. [PMID: 39633065 DOI: 10.1038/s41388-024-03246-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/19/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
Cervical cancer is the most common gynecologic cancer. Autophagy is involved in the progression of CCa. ULK1 is a crucial kinase in autophagy initiation. However, few studies have investigated the role of ULK1 phosphorylation at tyrosine residues in the progression of CCa, and the underlying mechanism remains elusive. In this study, we demonstrated that JAK2 is a novel upstream kinase that phosphorylates ULK1 at the tyrosine site. JAK2 interacts with and phosphorylates ULK1 at Tyr1007. The phosphorylation of ULK1 at Y1007 increases its activity and stability, activates autophagy, and promotes the progression of CCa. We further showed that the phosphorylation of ULK1 at Y1007 is a predictive marker of CCa patient outcome. Furthermore, we identified SRPK1 as a potential downstream substrate of ULK1 to promote the progression of CCa. Our research sheds light on the molecular mechanism of CCa progression, through JAK2/ULK1 axis, and emphasizes the phosphorylation of ULK1 at Y1007 as a predictor of CCa.
Collapse
Affiliation(s)
- Qiuhong Duan
- Translational Medical Center, Huaihe Hospital, Henan University, Kaifeng, Henan, 475000, PR China.
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China.
- Medical and industry crossover Research Institute of Medical college, Henan University, Kaifeng, Henan, 475000, PR China.
| | - Wei Wang
- Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430033, PR China
| | - Hua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Juanjuan Xiao
- Translational Medical Center, Huaihe Hospital, Henan University, Kaifeng, Henan, 475000, PR China
| | - Han Xiao
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430015, PR China.
| | - Feng Zhu
- Translational Medical Center, Huaihe Hospital, Henan University, Kaifeng, Henan, 475000, PR China.
- Medical and industry crossover Research Institute of Medical college, Henan University, Kaifeng, Henan, 475000, PR China.
| | - Hui Lu
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430015, PR China.
| |
Collapse
|
5
|
Liu K, Zhu J, Bao Y, Fang J, Zhou S, Fan J. Proliferation and migration inhibition of adenoid cystic carcinoma cells through autophagy suppression via GLUT1 knockdown. Heliyon 2025; 11:e42894. [PMID: 40070961 PMCID: PMC11894374 DOI: 10.1016/j.heliyon.2025.e42894] [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: 09/18/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Multiple studies have demonstrated a significant association between glucose transporter-1 (GLUT1) and the development and recurrence of adenoid cystic carcinoma (ACC). In this study, we investigated the impact of GLUT1 knockdown on adenoid cystic carcinoma. Our findings revealed that hypoxic conditions promoted the progression and autophagy of SACC83 and SACC-LM cell lines, an effect that was mitigated by GLUT1 knockdown. In vivo experiments showed that the combination of lentivirus-delivered GLUT1 shRNA and autophagy inhibitor chloroquine (CQ) produced the most substantial reduction in tumor volume, weight, Ki67 expression, and autophagy in tumor tissues. In conclusion, hypoxia facilitates ACC progression by upregulating GLUT1 expression. The suppression of GLUT1 expression and autophagy effectively inhibited ACC cell proliferation both in vitro and in vivo.
Collapse
Affiliation(s)
- Kan Liu
- Department of Otolaryngology, Zhejiang Sian International Hospital of Jiaxing City, Jiaxing City, Zhejiang Province, China
| | - Jinlong Zhu
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yangyang Bao
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jin Fang
- Department of Otolaryngology, Zhejiang Sian International Hospital of Jiaxing City, Jiaxing City, Zhejiang Province, China
| | - Shuihong Zhou
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jun Fan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Yang T, Fan Y, Bai G, Huang Y. RFC4 confers radioresistance of esophagus squamous cell carcinoma through regulating DNA damage response. Am J Physiol Cell Physiol 2025; 328:C367-C380. [PMID: 39672175 DOI: 10.1152/ajpcell.00533.2024] [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: 07/31/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 12/15/2024]
Abstract
Radioresistance in esophageal squamous cell carcinoma (ESCC) is a critical factor leading to treatment failure and recurrence, yet its underlying molecular mechanisms remain unclear. This study aimed to investigate the role of replication factor C4 (RFC4) in ESCC radioresistance and to explore the underlying mechanisms. We utilized online bioinformatics tools to analyze the properties, functions, and prognostic significance of RFC4 in ESCC. We established cell lines with varying RFC4 expression levels and subjected them to radiation exposure. RFC4 expression was assessed using quantitative real-time polymerase chain reaction (qRT-PCR), immunohistochemistry, and immunoblotting. Cell proliferation was evaluated with MTT, 5-ethynyl-2'-deoxyuridine (EdU), and colony formation assays. Apoptosis and cell cycle distribution were analyzed by flow cytometry. Western blotting and immunofluorescence were used to study the impact of RFC4 on the DNA damage response in ESCC cells. A xenograft mouse model was employed to assess tumor growth in vivo. RFC4 expression was significantly upregulated in ESCC tissues and cells, particularly in radioresistant cases. Functional experiments revealed that RFC4 promotes cell proliferation, inhibits apoptosis, induces cell cycle arrest, and mitigates radiation-induced DNA damage responses. Mechanistically, RFC4-mediated radioresistance in ESCC may involve the inactivation of the p53 signaling pathway. In animal studies, RFC4 knockdown, either alone or in combination with radiation therapy, effectively suppressed the growth of xenograft tumors. These findings highlight the potential of targeting RFC4 to overcome radioresistance by modulating the DNA damage response in ESCC, offering promising therapeutic avenues for patients with ESCC.NEW & NOTEWORTHY Our research indicates that replication factor C4 (RFC4) plays a role in conferring radioresistance to esophageal squamous cell carcinoma (ESCC) by bolstering DNA damage repair, primarily through the inhibition of the p53 signaling pathway. This finding positions RFC4 as a promising therapeutic target for combating radioresistance in ESCC, although further research is required to fully comprehend its intricate role in the disease.
Collapse
Affiliation(s)
- Tao Yang
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yue Fan
- Discipline Construction Office, Baoan Central Hospital of Shenzhen, Shenzhen, China
| | - Guang Bai
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yinpeng Huang
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
7
|
Luo Y, Liu R, Zhang H, Wang H, Yin H, Tian G, Wang B, Yan Y, Ding Z, Dai J, Niu L, Yuan G, Pan Y. Amantadine against glioma via ROS-mediated apoptosis and autophagy arrest. Cell Death Dis 2024; 15:834. [PMID: 39548081 PMCID: PMC11568115 DOI: 10.1038/s41419-024-07228-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: 05/07/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
Glioma is a common primary nervous system malignant tumor with poor overall cure rate and low survival rate, yet successful treatment still remains a challenge. Here, we demonstrated that amantadine (AMT) exhibits the powerful anti-glioma effect by promoting apoptosis and autophagy in vivo and in vitro. Mechanistically, amantadine induces a large amount of reactive oxygen species (ROS) accumulation in glioma cells, and then triggers apoptosis by destroying mitochondria. In addition, amantadine induces the initiation of autophagy and inhibits the fusion of autophagosome and lysosome, consequently performing an anti-glioma role. Taken together, our findings suggest that amantadine could be a promising anti-glioma drug that inhibits glioma cells by inducing apoptosis and autophagy, which may provide a novel potential treatment option for patients.
Collapse
Affiliation(s)
- Yusong Luo
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Ruolan Liu
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - He Zhang
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Hongyu Wang
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Hang Yin
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Guopeng Tian
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Bo Wang
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Yunji Yan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Zilin Ding
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Junqiang Dai
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Liang Niu
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Guoqiang Yuan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
| | - Yawen Pan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
- Academician Workstation, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
| |
Collapse
|
8
|
Xu X, Miao C, Yang S, Xiao L, Gao Y, Wu F, Xu J. Investigating potential drug targets for IgA nephropathy and membranous nephropathy through multi-queue plasma protein analysis: a Mendelian randomization study based on SMR and co-localization analysis. BioData Min 2024; 17:49. [PMID: 39516845 PMCID: PMC11545554 DOI: 10.1186/s13040-024-00405-w] [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: 07/27/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Membranous nephropathy (MN) and IgA nephropathy (IgAN) pose challenges in clinical treatment with existing therapies primarily focusing on symptom relief and often yielding unsatisfactory outcomes. The search for novel drug targets remains crucial to address the shortcomings in managing both kidney diseases. METHODS Utilizing GWAS data for MN (ncase = 2150, ncontrol = 5829) and IgAN (ncase = 15587, ncontrol = 462197), instrumental variables for plasma proteins were derived from recent GWAS. Sensitivity analysis involved bidirectional Mendelian randomization analysis, MR Steiger, Bayesian co-localization, and Phenotype scanning. The SMR analysis using eQTL data from the eQTLGen Consortium was conducted to assess the availability of selected protein targets. The PPI network was constructed to reveal potential associations with existing drug treatment targets. RESULTS The study, subjected to the stringent Bonferroni correction, revealed significant associations: four proteins with MN and three proteins with IgAN. In plasma protein cis-pQTL data from two cohorts, an increase in one standard deviation in PLA2R1 (OR = 2.01, 95%CI = 1.83-2.21), AIF1 (OR = 9.04, 95%CI = 4.69-17.41), MLN (OR = 3.79, 95%CI = 2.12-6.78), and NFKB1 (OR = 29.43, 95%CI = 7.73-112.0) was associated with an increased risk of MN. Additionally, in plasma protein cis-pQTL data, a standard deviation increase in FCGR3B (OR = 1.15, 95%CI = 1.09-1.22) and BTN3A1 (OR = 4.05, 95%CI = 2.65-6.19) correlated with elevated IgAN risk, while AIF1 (OR = 0.58, 95%CI = 0.46-0.73) exhibited IgAN protection. Bayesian co-localization indicated that PLA2R1 (coloc.abf-PPH4 = 0.695), NFKB1 (coloc.abf-PPH4 = 0.949), FCGR3B (coloc.abf-PPH4 = 0.909), and BTN3A1 (coloc.abf-PPH4 = 0.685) share the same variants associated with MN and IgAN. The SMR analysis indicated a causal link between NFKB1 and BTN3A1 plasma protein eQTL in both conditions, and BTN3A1 was validated externally. CONCLUSION Genetically influenced plasma levels of PLA2R1 and NFKB1 impact MN risk, while FCGR3B and BTN3A1 levels are causally linked to IgAN risk, suggesting potential drug targets for further clinical exploration, notably BTN3A1 for IgAN.
Collapse
Affiliation(s)
- Xinyi Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Changhong Miao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shirui Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Lu Xiao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Ying Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Fangying Wu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jianbo Xu
- Department of Clinical Laboratory, Jinhua Maternal and Child Health Care Hospital, Jinhua, Zhejiang, 321000, China.
| |
Collapse
|
9
|
Kone AS, Ghouzlani A, Qandouci A, Issam Salah NEI, Bakoukou Y, Lakhdar A, Karkouri M, Badou A. High expression of BTN3A1 is associated with clinical and immunological characteristics and predicts a poor prognosis in advanced human gliomas. Front Immunol 2024; 15:1397486. [PMID: 38863709 PMCID: PMC11165028 DOI: 10.3389/fimmu.2024.1397486] [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: 03/07/2024] [Accepted: 05/13/2024] [Indexed: 06/13/2024] Open
Abstract
Introduction Gliomas represent the most prevalent and aggressive tumors within the central nervous system. Despite the current standard treatments, the median survival time for glioblastoma patients remains dismal, hovering around 14 months. While attempts have been made to inhibit the PD-1/PD-L1 and CTLA-4/CD80-CD86 axes through immunotherapy, the outcomes have yet to demonstrate significant efficacy. The immune checkpoint Butyrophilin 3A1 (BTN3A1) can either be involved in advantageous or detrimental function depending on the cancer type. Methods In our study, we utilized a Moroccan cohort to delve into the role of BTN3A1 in gliomas. A transcriptomic analysis was conducted on 34 patients, which was then corroborated through a protein analysis in 27 patients and validated using the TCGA database (n = 667). Results Our results revealed an elevated expression of BTN3A1 in glioblastoma (grade 4), as evidenced in both the TCGA database and our cohort of Moroccan glioma patients. Within the TCGA cohort, BTN3A1 expression was notably higher in patients with wild-type IDH. We observed a positive correlation between BTN3A1 expression and immune infiltration of B cells, CD8+ T cells, naive CD4+ T cells, and M2 macrophages. Patients exhibiting increased BTN3A1 expression also presented elevated levels of TGF-β, IL-10, and TIM-3 compared to those with reduced BTN3A1 expression. Notably, patients with high BTN3A1 expression were associated with a poorer prognosis than their counterparts with lower expression. Conclussion Our findings suggest that BTN3A1 might promote the establishment of an immunosuppressive microenvironment. Consequently, targeting BTN3A1 could offer novel therapeutic avenues for the management of advanced gliomas.
Collapse
Affiliation(s)
- Abdou-samad Kone
- Immuno-Genetics and Human Pathology Laboratory (LIGEP), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Amina Ghouzlani
- Immuno-Genetics and Human Pathology Laboratory (LIGEP), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Ahmed Qandouci
- Immuno-Genetics and Human Pathology Laboratory (LIGEP), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Nour el Imane Issam Salah
- Immuno-Genetics and Human Pathology Laboratory (LIGEP), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Yann Bakoukou
- Immuno-Genetics and Human Pathology Laboratory (LIGEP), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Abdelhakim Lakhdar
- Department of Neurosurgery, University Hospital Center (UHC) Ibn Rochd, Casablanca, Morocco
| | - Mehdi Karkouri
- Laboratory of Pathological Anatomy, University Hospital Center (CHU) Ibn Rochd, Hassan II University, Casablanca, Morocco
| | - Abdallah Badou
- Immuno-Genetics and Human Pathology Laboratory (LIGEP), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Mohammed VI Center for Research and Innovation, Rabat, Morocco and Mohammed VI University of Sciences and Health, Casablanca, Morocco
| |
Collapse
|
10
|
Jung YY, Ahn KS, Shen M. Unveiling autophagy complexity in leukemia: The molecular landscape and possible interactions with apoptosis and ferroptosis. Cancer Lett 2024; 582:216518. [PMID: 38043785 DOI: 10.1016/j.canlet.2023.216518] [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: 11/03/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/05/2023]
Abstract
Autophagy is a self-digestion multistep process in which causes the homeostasis through degradation of macromolecules and damaged organelles. The autophagy-mediated tumor progression regulation has been a critical point in recent years, revealing the function of this process in reduction or acceleration of carcinogenesis. Leukemia is a haematological malignancy in which abnormal expansion of hematopoietic cells occurs. The current and conventional therapies from chemotherapy to cell transplantation have failed to appropriately treat the leukemia patients. Among the mechanisms dysregulated in leukemia, autophagy is a prominent one in which can regulate the hallmarks of this tumor. The protective autophagy inhibits apoptosis and ferroptosis in leukemia, while toxic autophagy accelerates cell death. The proliferation and invasion of tumor cells are tightly regulated by the autophagy. The direction of regulation depends on the function of autophagy that is protective or lethal. The protective autophagy accelerates chemoresistance and radio-resistsance. The non-coding RNAs, histone transferases and other pathways such as PI3K/Akt/mTOR are among the regulators of autophagy in leukemia progression. The pharmacological intervention for the inhibition or induction of autophagy by the compounds including sesamine, tanshinone IIA and other synthetic compounds can chance progression of leukemia.
Collapse
Affiliation(s)
- Young Yun Jung
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | - Mingzhi Shen
- Department of General Medicine, Hainan Hospital of PLA General Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Sanya, China.
| |
Collapse
|
11
|
Hill RM, Fok M, Grundy G, Parsons JL, Rocha S. The role of autophagy in hypoxia-induced radioresistance. Radiother Oncol 2023; 189:109951. [PMID: 37838322 PMCID: PMC11046710 DOI: 10.1016/j.radonc.2023.109951] [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: 06/21/2023] [Revised: 09/25/2023] [Accepted: 09/29/2023] [Indexed: 10/16/2023]
Abstract
Radiotherapy is a widely used treatment modality against cancer, and although survival rates are increasing, radioresistant properties of tumours remain a significant barrier for curative treatment. Tumour hypoxia is one of the main contributors to radioresistance and is common in most solid tumours. Hypoxia is responsible for many molecular changes within the cell which helps tumours to survive under such challenging conditions. These hypoxia-induced molecular changes are predominantly coordinated by the hypoxia inducible factor (HIF) and have been linked with the ability to confer resistance to radiation-induced cell death. To overcome this obstacle research has been directed towards autophagy, a cellular process involved in self degradation and recycling of macromolecules, as HIF plays a large role in its coordination under hypoxic conditions. The role that autophagy has following radiotherapy treatment is conflicted with evidence of both cytoprotective and cytotoxic effects. This literature review aims to explore the intricate relationship between radiotherapy, hypoxia, and autophagy in the context of cancer treatment. It provides valuable insights into the potential of targeting autophagy as a therapeutic strategy to improve the response of hypoxic tumours to radiotherapy.
Collapse
Affiliation(s)
- Rhianna Mae Hill
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, L7 8TX, UK
| | - Matthew Fok
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, L7 8TX, UK
| | - Gabrielle Grundy
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, L7 8TX, UK
| | - Jason Luke Parsons
- Institute of Cancer and Genomic Sciences, University of Birmingham, B15 2TT, UK
| | - Sonia Rocha
- Department of Biochemistry and Systems Biology, University of Liverpool, L69 7ZB, UK.
| |
Collapse
|
12
|
Wang M, Gao M, Chen Y, Wu J, Wang X, Shu Y. PLCD3 promotes malignant cell behaviors in esophageal squamous cell carcinoma via the PI3K/AKT/P21 signaling. BMC Cancer 2023; 23:921. [PMID: 37773107 PMCID: PMC10542242 DOI: 10.1186/s12885-023-11409-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: 04/05/2023] [Accepted: 09/16/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Phospholipase C Delta 3 (PLCD3) is a member of phospholipase C(PLC) Protein and PLCD3 protein plays a prominent role in many cancers. However, little is known about the role of PLCD3 in esophageal squamous cell carcinoma (ESCC). MATERIAL AND METHODS We analyzed PLCD3 mRNA and protein expression in ESCC tissues and cell lines by immunohistochemistry, quantitative real-time PCR, and western blot. The correlation between PLCD3 expression and clinicopathological characteristics was also analyzed. CCK8, colony formation, wound-healing, and transwell assays were conducted to measure cell functional alternations. Flow cytometry was performed to assess the apoptosis rate and cell cycle caused by PLCD3 knockdown. Xenograft models in nude mice to clarify the role of PLCD3 in ESCC. Key proteins in the PI3K / AKT signaling pathway after treatment of ECA109 and KYSE150 cells with the AKT inhibitor MK2206 were analyzed by western blot. RESULTS PLCD3 was highly expressed in ESCC tissues and cell lines. PLCD3 expression levels correlated with pathologic stage and lymphatic metastasis. PLCD3 knockdown inhibited cell proliferation, migration, invasion, promoted apoptosis, and caused the cell cycle arrest in the G1 phase. PLCD3 overexpression promoted cell proliferation, migration, and invasion. In vivo experiments with xenografts demonstrated that PLCD3 promoted ESCC tumorigenesis. Finally, Overexpression of PLCD3 activated the PI3K / AKT / P21 signaling. CONCLUSION PLCD3 promotes malignant cell behaviors in esophageal squamous cell carcinoma via the PI3K/AKT/P21 signaling and could serve as a potential target for ESCC treatment.
Collapse
Affiliation(s)
| | - Mingjun Gao
- Dalian Medical University, Dalian, 116000, China
| | - Yong Chen
- Dalian Medical University, Dalian, 116000, China
| | - Jun Wu
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, China
| | - Xiaolin Wang
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital, No. 98 Nantong West Road, Yangzhou, 225000, China.
| | - Yusheng Shu
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital, No. 98 Nantong West Road, Yangzhou, 225000, China.
| |
Collapse
|
13
|
An L, Li M, Jia Q. Mechanisms of radiotherapy resistance and radiosensitization strategies for esophageal squamous cell carcinoma. Mol Cancer 2023; 22:140. [PMID: 37598158 PMCID: PMC10439611 DOI: 10.1186/s12943-023-01839-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/02/2023] [Indexed: 08/21/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the sixth most common cause of cancer-related mortality worldwide, with more than half of them occurred in China. Radiotherapy (RT) has been widely used for treating ESCC. However, radiation-induced DNA damage response (DDR) can promote the release of cytokines and chemokines, and triggers inflammatory reactions and changes in the tumor microenvironment (TME), thereby inhibiting the immune function and causing the invasion and metastasis of ESCC. Radioresistance is the major cause of disease progression and mortality in cancer, and it is associated with heterogeneity. Therefore, a better understanding of the radioresistance mechanisms may generate more reversal strategies to improve the cure rates and survival periods of ESCC patients. We mainly summarized the possible mechanisms of radioresistance in order to reveal new targets for ESCC therapy. Then we summarized and compared the current strategies to reverse radioresistance.
Collapse
Affiliation(s)
- Lingbo An
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- College of Medical Technology, Xi'an Medical University, Xi'an, China
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| | - Qingge Jia
- Department of Reproductive Medicine, Xi'an International Medical Center Hospital, Northwest University, Xi'an, China.
| |
Collapse
|
14
|
Zlatareva I, Wu Y. Local γδ T cells: translating promise to practice in cancer immunotherapy. Br J Cancer 2023; 129:393-405. [PMID: 37311978 PMCID: PMC10403623 DOI: 10.1038/s41416-023-02303-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/06/2023] [Accepted: 05/15/2023] [Indexed: 06/15/2023] Open
Abstract
Rapid bench-to-bedside translation of basic immunology to cancer immunotherapy has revolutionised the clinical practice of oncology over the last decade. Immune checkpoint inhibitors targeting αβ T cells now offer durable remissions and even cures for some patients with hitherto treatment-refractory metastatic cancers. Unfortunately, these treatments only benefit a minority of patients and efforts to improve efficacy through combination therapies utilising αβ T cells have seen diminishing returns. Alongside αβ T cells and B cells, γδ T cells are a third lineage of adaptive lymphocytes. Less is known about these cells, and they remain relatively untested in cancer immunotherapy. Whilst preclinical evidence supports their utility, the few early-phase trials involving γδ T cells have failed to demonstrate convincing efficacy in solid cancers. Here we review recent progress in our understanding of how these cells are regulated, especially locally within tissues, and the potential for translation. In particular, we focus on the latest advances in the field of butyrophilin (BTN) and BTN-like (BTNL) regulation of γδ T cells and speculate on how these advances may address the limitations of historical approaches in utilising these cells, as well as how they may inform novel approaches in deploying these cells for cancer immunotherapy.
Collapse
Affiliation(s)
- Iva Zlatareva
- Peter Gorer Department of Immunobiology, King's College London, London, SE1 9RT, UK
| | - Yin Wu
- Peter Gorer Department of Immunobiology, King's College London, London, SE1 9RT, UK.
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London, SE1 9RT, UK.
- Department of Medical Oncology, Guy's Hospital, London, SE1 9RT, UK.
| |
Collapse
|
15
|
Zeng L, Zheng W, Liu X, Zhou Y, Jin X, Xiao Y, Bai Y, Pan Y, Zhang J, Shao C. SDC1-TGM2-FLOT1-BHMT complex determines radiosensitivity of glioblastoma by influencing the fusion of autophagosomes with lysosomes. Theranostics 2023; 13:3725-3743. [PMID: 37441590 PMCID: PMC10334832 DOI: 10.7150/thno.81999] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 06/07/2023] [Indexed: 07/15/2023] Open
Abstract
Rationale: Glioblastoma (GBM) is the most common and malignant primary brain tumor in adults. Radiotherapy has long been an important treatment for GBM. Despite recent advances in tumor radiotherapy, the prognosis of GBM remains poor due to radioresistance. Autophagy has been reported as a basic factor to prolong the survival of tumor under radiation stress, but the molecular mechanism of how autophagy contributes to GBM radioresistance was still lacking. Methods: We established radioresistant GBM cells and identified their protein profiles by Tandem mass tag (TMT) quantitative proteomic analysis, then chose the radioresistant genes based on the TMT analysis of GBM cells and differentially expressed genes (DEGs) analysis of GEO database. Colony formation, flow cytometry, qPCR, western blotting, mRFP-GFP-LC3, transmission electron microscopy, immunofluorescence, and co-IP assays were conducted to investigate the regulation mechanisms among these new-found molecules. Results: Syndecan 1 (SDC1) and Transglutaminase 2 (TGM2) were both overexpressed in the radioresistant GBM cells and tissues, contributing to the dismal prognosis of radiotherapy. Mechanically, after irradiation, SDC1 carried TGM2 from cell membrane into cytoplasm, and transported to lysosomes by binding to flotillin 1 (FLOT1), then TGM2 recognized the betaine homocysteine methyltransferase (BHMT) on autophagosomes to coordinate the encounter between autophagosomes and lysosomes. Conclusions: The SDC1-TGM2-FLOT1-BHMT copolymer, a novel member of the protein complexes involved in the fusion of lysosomes and autophagosomes, maintained the autophagic flux in the irradiated tumor cells and ultimately enhanced radioresistance of GBM, which provides new insights of the molecular mechanism and therapeutic targets of radioresistant GBM.
Collapse
Affiliation(s)
- Liang Zeng
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wang Zheng
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210009, China
| | - Xinglong Liu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yuchuan Zhou
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaoya Jin
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yuqi Xiao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yang Bai
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yan Pan
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jianghong Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|