1
|
Arslan S, Berber I, Kuku I, Kaya E, Erkurt MA, Biçim S, Kaya A, Pinar A. When should I use filgrastim after autologous transplantation in MM patients? Transfus Apher Sci 2025; 64:104171. [PMID: 40513502 DOI: 10.1016/j.transci.2025.104171] [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: 01/06/2025] [Revised: 05/31/2025] [Accepted: 06/04/2025] [Indexed: 06/16/2025]
Abstract
BACKGROUND Granulocyte colony-stimulating factor (G-CSF) is routinely administered following autologous stem cell transplantation in patients with multiple myeloma (MM); however, the optimal timing for its initiation remains unclear. While previous studies have evaluated heterogeneous patient cohorts, including those with MM, Non-Hodgkin's Lymphoma, and Hodgkin's Lymphoma, this study focuses exclusively on MM patients. We aimed to compare the outcomes of initiating G-CSF either on day + 1 post-transplantation or upon the onset of neutropenia, with particular emphasis on neutrophil and platelet engraftment times, to help define an optimal G-CSF administration strategy in this patient population. STUDY DESIGN AND METHODS This retrospective study included 122 MM patients who underwent autologous hematopoietic stem cell transplantation between 2016 and 2022 at the Hematology Clinic of İnönü University Turgut Özal Medical Center. Patients were evenly divided into two groups. In Group 1, filgrastim was initiated on day + 1 post-transplantation, while in Group 2, it was administered after the onset of neutropenia. Neutrophil and platelet engraftment times, as well as antibiotic usage, were compared between the groups. RESULTS There were no statistically significant differences in neutrophil or platelet engraftment times or in antibiotic usage between the two groups (p > 0.05). The median neutrophil and platelet engraftment times were 14 and 15 days, respectively, in the day + 1 group, and 15 and 14 days in the neutropenia-guided group. However, the median number of filgrastim injections was significantly lower in the neutropenia group (8 injections, range: 6-12) compared to the day + 1 group (14 injections, range: 8-24) (p < 0.001). CONCLUSION Initiating G-CSF upon the development of neutropenia is as effective as early (day +1) administration in MM patients undergoing autologous transplantation. This delayed strategy does not adversely affect engraftment or antibiotic requirements and significantly reduces the number of G-CSF injections, offering potential benefits in terms of cost-effectiveness and reduced side effects.
Collapse
Affiliation(s)
- Süleyman Arslan
- Turgut Ozal Medical Center, Departmentof Hematology, Inonu University, Malatya, Turkey.
| | - Ilhami Berber
- Turgut Ozal Medical Center, Departmentof Hematology, Inonu University, Malatya, Turkey.
| | - Irfan Kuku
- Turgut Ozal Medical Center, Departmentof Hematology, Inonu University, Malatya, Turkey.
| | - Emin Kaya
- Turgut Ozal Medical Center, Departmentof Hematology, Inonu University, Malatya, Turkey.
| | - Mehmet Ali Erkurt
- Turgut Ozal Medical Center, Departmentof Hematology, Inonu University, Malatya, Turkey.
| | - Soykan Biçim
- Turgut Ozal Medical Center, Departmentof Hematology, Inonu University, Malatya, Turkey.
| | - Ahmet Kaya
- Turgut Ozal Medical Center, Departmentof Hematology, Inonu University, Malatya, Turkey.
| | - Abdulvahap Pinar
- Faculty of Medicine, Department of Biostatistics and Medical Informatics, Adıyaman, University, Adıyaman, Turkey.
| |
Collapse
|
2
|
Chen W, Shi K, Mo D, Pan M, Bei ZW, Deng HZ, Yang PP, Tong Q, Yuan LP, Wan YY, Liu JF, Pan LL, Qian ZY. Development of orthotopic mouse models for mid-low rectal cancer. Acta Pharmacol Sin 2025; 46:1772-1781. [PMID: 39939805 PMCID: PMC12098718 DOI: 10.1038/s41401-025-01489-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/17/2025] [Indexed: 02/14/2025]
Abstract
Mid-low rectal cancer is one of the most common types of rectal cancer and has a poor prognosis. Surgery and chemoradiotherapy are the main treatments for early and advanced rectal cancer with an overall 5-year relative survival rate of only 56.9%. Development of novel antitumor agents is needed. Animal models of disease are indispensable for drug development. The most commonly used animal models of rectal cancer are established by inducing tumors by the subcutaneous transplantation, cecum or peritoneal injection, but not injection in the rectum. Their tumor microenvironment differs from that of rectal tumors in situ, which is hard to precisely simulate the occurrence and development process and drug response of human rectal cancer. In this study, we established orthotopic mouse models of mid-low rectal cancer with primary tumors originating from the rectum, including two models that could simulate the early and advanced stages of the disease, respectively. In the first model, the local primary tumor was restricted to the rectal area of the anal verge by rectal submucosal injection, its growth could be monitored with IVIS live imaging and magnetic resonance imaging. Histological analysis confirmed that the tumor originated from the submucosal layer and then invaded the muscular layer without metastatic tumors. This model may be useful for evaluating drugs for early mid-low rectal cancer in the future. The second model featuring a rectal primary tumor accompanied with abdominal metastases was established via rectal serosal injection. In this model, a large tumor formed at the rectal injection site and then metastasized to the abdominal cavity, reproducing the process from occurrence to metastasis of mid-low rectal cancer, and may be a good tool for the evaluation of drugs for advanced-stage disease. The injection methods used in these models do not require the aid of special colonoscopes, are simple and easy to operate, and have high tumor tumorigenicity and reproducibility. These results suggest that our staged modeling can provide targeted choices for preclinical drug research of mid-low rectal cancer at different stages.
Collapse
Affiliation(s)
- Wen Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kun Shi
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dong Mo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Pan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhong-Wu Bei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Han-Zhi Deng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Pei-Pei Yang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qi Tong
- Department of Cardiovascular Surgery, West China Hospital, Med-X Center for Materials, Sichuan University, Chengdu, 610041, China
| | - Li-Ping Yuan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yi-Yao Wan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jia-Feng Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li-Li Pan
- Department of Nuclear Medicine and Clinical Nuclear Medicine Research Lab, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhi-Yong Qian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
3
|
Li Y, Wang Q. Myoepithelial carcinoma of submaxillary gland: A case report. Oncol Lett 2025; 29:261. [PMID: 40230425 PMCID: PMC11995683 DOI: 10.3892/ol.2025.15007] [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: 09/17/2024] [Accepted: 10/29/2024] [Indexed: 04/16/2025] Open
Abstract
Myoepithelial carcinoma (MEC) is a malignant tumour composed almost entirely of cells differentiated from the myoepithelium. It is rare, most commonly occurring in the parotid gland. In the current study, a rare case of MEC in the submandibular gland was presented. An 82-year-old male patient was admitted with a swelling in the upper left neck that had been present for 60 years. Magnetic resonance imaging indicated a tumour in the region of the left submandibular gland, which was not easy to clearly distinguish from the gland itself and an enhancement scan was recommended but not performed. Furthermore, multiple lymph nodes of varying sizes were present in the submandibular region of the neck on both sides. These enlarged lymph nodes had signs of involvement. Accordingly, a surgery was performed. The pathological diagnosis indicated the presence of a malignant tumour in the left upper neck. The tumour was most likely an MEC with pleomorphic adenoma, based on the results of immunohistochemistry. The size of the tumour mass was 5×5×4 cm. Histology indicated that the tumour cells exhibited infiltrative growth, with a sparse arrangement and a predominance of hyaline and spindle-shaped cells. In certain regions, the tumour tissue displayed characteristics similar to pleomorphic adenoma. The results of the immunohistochemical analysis were as follows: CD31(-), CD34(-), cytokeratin (CK)7 (+), vimentin (+), Ki-67 (+, 70%), erythroid growth factor receptor (-), desmin (-), anaplastic lymphoma kinase (-), P40 (+), CK pan-antibody (+) and epithelial membrane antigen (-). The patient underwent a surgical procedure to remove the lesion in the neck under sedation with complex anaesthesia. The sample excised from the surgery was sent to the pathology department for diagnosis. Given the high recurrence frequency of MEC, monitoring patients closely after surgery is crucial.
Collapse
Affiliation(s)
- Yuetian Li
- Clinical School of Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Quanyi Wang
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| |
Collapse
|
4
|
Nomenclature for Factors of the HLA System, Update January, February and March 2025. Int J Immunogenet 2025; 52:162-187. [PMID: 40292998 PMCID: PMC12087755 DOI: 10.1111/iji.12713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
|
5
|
Wu SQ, Li XF, Qiu ZJ, Zhu ZJ, Chen XL, Chen P, Yuan XH, Zhan R, Li NN. Comparison of tandem and single autologous stem cell transplantation in multiple myeloma: a retrospective propensity score-matching study. BLOOD SCIENCE 2025; 7:e00235. [PMID: 40356608 PMCID: PMC12068754 DOI: 10.1097/bs9.0000000000000235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Affiliation(s)
- Shun-Quan Wu
- Department of Hematology, Fujian Medical University Union Hospital, Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fuzhou, China
| | - Xiao-Fan Li
- Department of Hematology, Fujian Medical University Union Hospital, Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fuzhou, China
- Translational Medicine Center on Hematology, Fujian Medical University, Fuzhou, China
| | - Zong-Jian Qiu
- Department of Hematology, Fujian Medical University Union Hospital, Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fuzhou, China
| | - Zhi-Juan Zhu
- Department of Hematology, Fujian Medical University Union Hospital, Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fuzhou, China
| | - Xian-Ling Chen
- Department of Hematology, Fujian Medical University Union Hospital, Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fuzhou, China
| | - Ping Chen
- Department of Hematology, Fujian Medical University Union Hospital, Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fuzhou, China
| | - Xiao-Hong Yuan
- Department of Hematology, Fujian Medical University Union Hospital, Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fuzhou, China
| | - Rong Zhan
- Department of Hematology, Fujian Medical University Union Hospital, Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fuzhou, China
| | - Nai-Nong Li
- Department of Hematology, Fujian Medical University Union Hospital, Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fuzhou, China
- Translational Medicine Center on Hematology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
6
|
Huang M, Wang W, Wang R, Tian R. The prognostic value of pretreatment [ 18F]FDG PET/CT parameters in esophageal cancer: a meta-analysis. Eur Radiol 2025; 35:3396-3408. [PMID: 39570366 DOI: 10.1007/s00330-024-11207-3] [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: 06/20/2024] [Revised: 08/19/2024] [Accepted: 10/14/2024] [Indexed: 11/22/2024]
Abstract
OBJECTIVES This study aims to evaluate the prognostic implications of pretreatment [18F]FDG-PET metrics in esophageal cancer patients through a meta-analysis of the existing literature. METHODS We carefully searched electronic databases, including PubMed and Embase, from inception to April 1, 2024, to identify studies describing the prognostic value of pretreatment PET metrics for advanced esophageal cancer. Clinical endpoints examined were overall survival (OS), recurrence-free survival (RFS)/disease-free survival (DFS), and progression-free survival (PFS). Hazard ratios (HRs) for PFS and OS were taken directly from the original reports. RESULTS Forty-seven publications, including 5504 patients, were included in our analysis. OS and PFS were analyzed in 31 and nine studies, respectively, and DFS/RFS was analyzed in 16 studies. The comprehensive pooled analysis revealed significant associations between metabolic parameters derived from positron emission tomography (PET) imaging and clinical outcomes. Expressly, the pooled HR indicated that patients with higher SUVmax were significantly associated with poor PFS (HR: 1.06; 95% CI: 1.01-1.12, p = 0.011) and poor RFS/DFS (HR: 1.09; 95% CI: 1.02-1.18, p = 0.019). Patients with higher SUVmean were significantly associated with poorer OS (HR: 1.07; 95% CI: 1.01-1.14, p = 0.025). High MTV was significantly associated with inferior OS (HR: 1.02; 95% CI: 1.00-1.05, p = 0.049). High TLG was significantly associated with poorer RFS/DFS (HR: 2.02; 95% CI: 1.11-3.68, p = 0.022). CONCLUSION This study unveiled pretreatment FDG-derived parameters as valuable prognostic indicators in assessing esophageal cancer outcomes. Specifically, SUVmax is associated with PFS and RFS/DFS. SUVmean and MTV were correlated with OS, and TLG was only associated with RFS/DFS. KEY POINTS Question Inconsistent findings on the prognostic value of pretreatment [18F]FDG PET parameters in esophageal cancer require comprehensive analysis to clarify their role in outcome prediction. Findings Higher pretreatment [18F]FDG-PET metrics (SUVmax, SUVmean, MTV, TLG) are associated with poor survival outcomes, emphasizing their potential value in enhancing prognostic assessments for esophageal cancer. Clinical relevance This study highlights the prognostic significance of pretreatment [18F]FDG-PET metrics in esophageal cancer, providing valuable insights for patient outcome prediction and potentially guiding personalized treatment strategies.
Collapse
Affiliation(s)
- Mingxing Huang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Weichen Wang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Rang Wang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Rong Tian
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
7
|
Corica DA, Bell SD, Zhao L, Lawler NJ, Poirier MA, Miller PJ, Wakefield MR, Fang Y. The Era of Precision Medicine: Advancing Treatment Paradigms for Small Cell Lung Cancer. Cancers (Basel) 2025; 17:1847. [PMID: 40507328 PMCID: PMC12153792 DOI: 10.3390/cancers17111847] [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: 04/14/2025] [Revised: 05/29/2025] [Accepted: 05/29/2025] [Indexed: 06/16/2025] Open
Abstract
Small cell lung cancer (SCLC) remains a challenge prognostically. A clinically silent early stage and predilection for early metastasis leads to over half of patients presenting with metastatic disease at the time of diagnosis. Akin to many other cancers, once SCLC metastasizes, current therapies begin to lose their effectiveness. The future of SCLC rests in innovative treatments aimed at improving patient outcomes. Chemotherapy and radiation remain the backbone treatment for SCLC. Most patients diagnosed with SCLC begin treatment with combination chemotherapy consisting of a platinum analog and topoisomerase inhibitor with or without concurrent radiation. Disease progression or recurrence warrants new treatment approaches. New chemotherapy combinations and advances in radiation precision offer patients novel approaches using the same backbone of treatment used in many other cancers. The introduction of newer therapeutic approaches, such as immune checkpoint inhibitors, small molecule targeted therapies, bispecific antibodies, and antibody-drug conjugates offer a bright future for patients with SCLC who fail first-line therapy. This review will focus on advancing treatment paradigms for SCLC in the era of precision medicine. Such a study might be helpful for pulmonologists and oncologists to manage precisely patients with SCLC.
Collapse
Affiliation(s)
- Derek A. Corica
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
| | - Scott D. Bell
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
| | - Lei Zhao
- The Department of Respiratory Medicine, the 2nd People’s Hospital of Hefei and Hefei Hospital Affiliated to Anhui Medical University, Hefei 230002, China;
| | - Nicholas J. Lawler
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
| | - McKade A. Poirier
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
| | - Peyton J. Miller
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
| | - Mark R. Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA;
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA;
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| |
Collapse
|
8
|
Genc S, Cicek B. Examination of PDK1/AKT/mTOR transcription and exosomal mRNA levels in human glioblastoma cell line treated with a combination of temozolomide and hesperidin. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04137-4. [PMID: 40372477 DOI: 10.1007/s00210-025-04137-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 04/01/2025] [Indexed: 05/16/2025]
Abstract
The most malignant type of tumor in the brain is high-grade gliomas. Glioblastoma (GB), a grade 4 glioma, has the lowest 5-year survival rate and is associated with poor prognosis. An important signaling pathway involved in the pathogenesis of GB is the mammalian target of rapamycin (mTOR). Therefore, our study aimed to investigate how exosomes obtained from GB cells applied with different doses of hesperidin (HSP) affect miR- 9 and change the PDK1/AKT/mTOR pathway. For this purpose, T98G cells were treated with different doses (5, 10, 25, and 50 µg/mL) of HSP in combination with temozolomide (TMZ- 10 µg/mL). At the end of 24 h, cell viability, flow cytometry, and biochemical tests were performed. Additionally, exosomes were isolated from cells belonging to the control, TMZ, and high-concentration TMZ-HSP groups. miR- 9, PDK1, PTEN, AKT- 1, Bax, Bcl- 2, and Caspase 3 genes were expressed in both application groups and exosomes belonging to these groups. HSP was found to reduce the viability of GB cells significantly. The viability was significantly reduced, especially in the TMZ-HSP 50 µg/mL group. Depending on the dose, there was a significant increase in the LDH level and oxidative stress level. The apoptosis level was approximately 26% in the TMZ-HSP 50 µg/mL group. Along with all this, gene expressions changed at the exosomal level, and miR- 9 and miR- 146 levels increased. Similarly, it changed the expression of proteins related to the PDK1/AKT/mTOR signaling pathway at the exosomal level (p < 0.05). In conclusion, the TMZ-HSP combination showed anticancer effects in T98G cells, influenced exosome profiles, and appeared non-toxic and potentially beneficial to healthy cells, highlighting its potential therapeutic value.
Collapse
Affiliation(s)
- Sidika Genc
- Faculty of Medicine, Department of Medical Pharmacology, Bilecik Şeyh Edebali University, Bilecik, 11230, Turkey.
| | - Betul Cicek
- Faculty of Medicine, Department of Physiology, Erzincan Binali Yildirim University, Erzincan, 24100, Turkey
| |
Collapse
|
9
|
Marsh SGE. Nomenclature for Factors of the HLA System, Update January, February, and March 2025. HLA 2025; 105:e70203. [PMID: 40376765 DOI: 10.1111/tan.70203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 05/18/2025]
Affiliation(s)
- Steven G E Marsh
- UCL Cancer Institute, University College London (UCL), London, UK
| |
Collapse
|
10
|
Yuan XC, Jia P, Tian T, Zhu J, Zhang XY. Endoscopic submucosal dissection for esophageal precancerous lesions and early esophageal carcinoma: Analysis of efficacy and serum tumor markers. World J Gastrointest Surg 2025; 17:103700. [PMID: 40291880 PMCID: PMC12019039 DOI: 10.4240/wjgs.v17.i4.103700] [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: 01/08/2025] [Revised: 02/25/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Tumor progression in patients with esophageal precancerous lesions (EPLs) or early esophageal carcinoma (EEC) is typically confined in both extent and location. Prompt and effective intervention significantly improves treatment outcomes and prognosis for these individuals. AIM To determine the effect of endoscopic submucosal dissection (ESD) on efficacy, serum tumor markers (STMs), and 6-month postoperative recurrence rate in patients with either EPL or EEC. METHODS This study initially enrolled 120 patients with EPL or EEC, who were admitted from April 2021 to April 2024. Participants were divided into the control group (60 cases), which underwent thoracotomy, and the research group (60 cases) which received ESD treatment. The comparative analysis involved information regarding the efficacy (dissection area and resection rate per unit time), complications (delayed bleeding, wound infection, esophageal reflux, and postoperative esophageal stenosis), surgery-related parameters (bleeding volume, operation duration, and hospital length of stay), STMs [carcinoembryonic antigen (CEA), carbohydrate antigen 724 (CA724), and tumor-specific growth factor (TSGF)], and the 6-month postoperative recurrence rate of the two groups. RESULTS Data indicated statistically higher dissection area and resection rate per unit of time in the research group than in the control group. Meanwhile, the research group demonstrated a notably lower overall incidence rate of complications, bleeding volume, operation duration, and hospital length of stay. Further, the CEA, CA724, and TSGF were markedly reduced in the research group after treatment, which were statistically lower compared to the baseline and those of the control group. Finally, during the follow-up, a comparable 6-month postoperative recurrence rate was determined in the two groups. CONCLUSION ESD is clinically effective and safe for EPL and EEC and can significantly restore abnormally increased levels of STMs.
Collapse
Affiliation(s)
- Xiao-Chen Yuan
- Department of Gastroenterology, The Affiliated Taizhou Second People’s Hospital of Yangzhou University, Taizhou 225300, Jiangsu Province, China
| | - Ping Jia
- Department of Gastroenterology, The Affiliated Taizhou Second People’s Hospital of Yangzhou University, Taizhou 225300, Jiangsu Province, China
| | - Tian Tian
- Department of Laboratory Medicine, The Affiliated Taizhou Second People’s Hospital of Yangzhou University, Taizhou 225300, Jiangsu Province, China
| | - Jun Zhu
- Department of Gastroenterology, The Affiliated Taizhou Second People’s Hospital of Yangzhou University, Taizhou 225300, Jiangsu Province, China
| | - Xiao-Yan Zhang
- Department of Gastroenterology, The Affiliated Taizhou Second People’s Hospital of Yangzhou University, Taizhou 225300, Jiangsu Province, China
| |
Collapse
|
11
|
de Souza Barbosa I, Pilotto Heming C, Moura Neto V, Aran V. The Role of RAS in CNS Tumors: A Key Player or an Overlooked Oncogene? Int J Mol Sci 2025; 26:4104. [PMID: 40362343 PMCID: PMC12071703 DOI: 10.3390/ijms26094104] [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: 03/22/2025] [Revised: 04/18/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
This review examines the prevalence, molecular mechanisms, and clinical implications of RAS mutations in Central Nervous System (CNS) tumors, with a particular focus on glioblastoma. We summarize the current understanding of RAS-driven oncogenic pathways, their contribution to tumor progression, and potential therapeutic strategies targeting RAS and its downstream effectors. Although direct RAS mutations are rare in primary CNS tumors, alterations in RAS signaling, such as NF-1 loss and aberrant receptor tyrosine kinase activation, contribute to malignant progression. Furthermore, emerging evidence links RAS mutations to brain metastases, highlighting their significance in CNS oncology. We also discuss recent clinical trials investigating RAS-targeted therapies, including covalent inhibitors, MEK inhibitors, and novel combination approaches. Given the increasing recognition of RAS pathway alterations in CNS malignancies, further research is needed to elucidate their role in tumor biology and explore targeted therapeutic interventions.
Collapse
Affiliation(s)
- Isabel de Souza Barbosa
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro 20261-901, Brazil; (I.d.S.B.); (C.P.H.); (V.M.N.)
- Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-853, Brazil
| | - Carlos Pilotto Heming
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro 20261-901, Brazil; (I.d.S.B.); (C.P.H.); (V.M.N.)
- Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-853, Brazil
| | - Vivaldo Moura Neto
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro 20261-901, Brazil; (I.d.S.B.); (C.P.H.); (V.M.N.)
| | - Veronica Aran
- Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-853, Brazil
- Laboratório de Morfogênese Celular (LMC), Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-853, Brazil
| |
Collapse
|
12
|
Li Z, Fang L, Lv L, He X, Luo W, Guo D, Liu Z. Radiation-Induced Fibrosarcoma in the Stoma: A Case Report and Literature Review. Case Rep Surg 2025; 2025:5313214. [PMID: 40297646 PMCID: PMC12037238 DOI: 10.1155/cris/5313214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 02/17/2025] [Indexed: 04/30/2025] Open
Abstract
Fibrosarcoma is a rare malignant neoplasm consisting of fibroblasts with a large variety of collagen production. They usually involve deep soft tissues in extremities and trunk. However, fibrosarcoma can be seen in fields that received previous irradiation. Here, we report a case of parastomal fibrosarcoma after laparoscope-assisted extra-levator abdominal perineal resection (ELAPE) and colostomy. Prior to surgery, the patient underwent neoadjuvant chemoradiotherapy. The patient received extensive stomal lumpectomy and stoma reconstruction. The patient is free of local or distal recurrence for 1 year and died 4 years after diagnosis.
Collapse
Affiliation(s)
- Zhao Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Liang Fang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Liang Lv
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xinjia He
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wenqiang Luo
- Department of Emergency Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Dong Guo
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhen Liu
- Department of Emergency Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
13
|
Chen LN, Zhou H, Xi K, Cheng S, Liu Y, Fu Y, Ma X, Xu P, Ji SY, Wang WW, Shen DD, Zhang H, Shen Q, Chai R, Zhang M, Yang L, Han F, Mao C, Cai X, Zhang Y. Proton perception and activation of a proton-sensing GPCR. Mol Cell 2025; 85:1640-1657.e8. [PMID: 40215960 DOI: 10.1016/j.molcel.2025.02.030] [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: 10/21/2024] [Revised: 01/22/2025] [Accepted: 02/28/2025] [Indexed: 04/20/2025]
Abstract
Maintaining pH at cellular, tissular, and systemic levels is essential for human health. Proton-sensing GPCRs regulate physiological and pathological processes by sensing the extracellular acidity. However, the molecular mechanism of proton sensing and activation of these receptors remains elusive. Here, we present cryoelectron microscopy (cryo-EM) structures of human GPR4, a prototypical proton-sensing GPCR, in its inactive and active states. Our studies reveal that three extracellular histidine residues are crucial for proton sensing of human GPR4. The binding of protons induces substantial conformational changes in GPR4's ECLs, particularly in ECL2, which transforms from a helix-loop to a β-turn-β configuration. This transformation leads to the rearrangements of H-bond network and hydrophobic packing, relayed by non-canonical motifs to accommodate G proteins. Furthermore, the antagonist NE52-QQ57 hinders human GPR4 activation by preventing hydrophobic stacking rearrangement. Our findings provide a molecular framework for understanding the activation mechanism of a human proton-sensing GPCR, aiding future drug discovery.
Collapse
Affiliation(s)
- Li-Nan Chen
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hui Zhou
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Kun Xi
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shizhuo Cheng
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yongfeng Liu
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yifan Fu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiangyu Ma
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China; State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing 210096, China
| | - Ping Xu
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China; Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Su-Yu Ji
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wei-Wei Wang
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Dan-Dan Shen
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Huibing Zhang
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qingya Shen
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing 210096, China
| | - Min Zhang
- College of Computer Science and Technology, Zhejiang University, Hangzhou 310027, China
| | - Lin Yang
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| | - Chunyou Mao
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China; Center for Structural Pharmacology and Therapeutics Development, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
| | - Xiujun Cai
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou 310016, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou 310016, China.
| | - Yan Zhang
- Department of Pathology of Sir Run Run Shaw Hospital, Department of Pharmacology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
14
|
Wang Y, Xiong Y, Gu S, Wang W, Yang Y, Liu P. Clinical benefit loss in myeloma patients declining autologous stem cell transplantation: a real-world study. Discov Oncol 2025; 16:534. [PMID: 40238028 PMCID: PMC12003250 DOI: 10.1007/s12672-025-02356-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/09/2025] [Indexed: 04/18/2025] Open
Abstract
Autologous hematopoietic stem cell transplantation (ASCT) is a recommended treatment for multiple myeloma (MM). Currently, with multiple treatment alternatives, patients' prognosis has improved significantly compared to the pre-proteasome inhibitor period. However, this has raised deliberations on the value and timing of ASCT. Compared with Western countries, the proportion of Chinese patients undergoing ASCT is relatively low. Nevertheless, this situation allows us to observe the treatment outcomes of transplant-eligible (TE) patients who haven't received ASCT and explore the role of ASCT in patients with distinct clinicopathological features. This real-world analysis encompassed 1059 newly diagnosed MM patients from 2012 to 2022, among whom 480 were TE. These patients were categorized into the TE-ASCT group (158 received ASCT) and the TE-no-ASCT group (322 did not receive ASCT). Disease progression and treatment response are evaluated based on the definition of IMWG. We found that the progression-free survival (PFS) was significantly prolonged in TE-ASCT group compared to TE-no-ASCT group, but there was no significant difference in overall survival (OS). Further exploratory analyses revealed that ASCT conspicuously augmented the PFS of patients aged ≤ 60 years, featuring ISS stage II/III, 1q gain/amplification, and positive minimal residual disease (MRD). Nevertheless, for patients aged > 60 years, presenting with ISS stage I, high-risk cytogenetics, renal insufficiency, and negative MRD, ASCT did not confer PFS benefits. Our investigation furnishes evidence of the clinical superiority of ASCT for MM patients with disparate clinicopathological characteristics in the contemporary treatment era, laying a groundwork for individualized ASCT selection.
Collapse
Affiliation(s)
- Yue Wang
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Yaqin Xiong
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Shiyang Gu
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Wenjing Wang
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Yang Yang
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Peng Liu
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China.
| |
Collapse
|
15
|
Jiang J, Liu F, Cui D, Xu C, Chi J, Yan T, Guo F. Novel molecular mechanisms of immune evasion in hepatocellular carcinoma: NSUN2-mediated increase of SOAT2 RNA methylation. Cancer Commun (Lond) 2025. [PMID: 40227950 DOI: 10.1002/cac2.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a deadly malignancy known for its ability to evade immune surveillance. NOP2/Sun RNA methyltransferase family member 2 (NSUN2), an RNA methyltransferase involved in carcinogenesis, has been associated with immune evasion and energy metabolism reprogramming. This study aimed to examine the molecular mechanisms underlying the involvement of NSUN2 in immune evasion and metabolic reprogramming of HCC. METHODS Single-cell transcriptomic sequencing was applied to examine cellular composition changes, particularly immune cell dynamics, in HCC and adjacent normal tissues. Bulk RNA-seq and proteomics identified key genes and proteins. Methylation sequencing and methylated RNA immunoprecipitation (MeRIP) were carried out to characterize the role of NSUN2 in 5-methylcytosine (m5C) modification of sterol O-acyltransferase 2 (SOAT2). Clinical samples from 30 HCC patients were analyzed using reverse transcription-quantitative polymerase chain reaction and Western blotting. Gene expression was manipulated using CRISPR/Cas9 and lentiviral vectors. In vitro co-culture models and metabolomics were used to study HCC cell-T cell interactions, energy metabolism, and immune evasion. Tumor growth in an orthotopic mouse model was monitored by bioluminescence imaging, with subsequent measurements of tumor weight, volume, and immunohistochemical staining. RESULTS Single-cell transcriptomic analysis identified a marked increase in malignant cells in HCC tissues. Cell communication analysis indicated that tumor cells might promote cancer progression by evading immune clearance. Multi-omics analyses identified NSUN2 as a key regulator in HCC development. MeRIP confirmed that NSUN2 facilitated the m5C modification of SOAT2. Analysis of human HCC tissue samples demonstrated pronounced upregulation of NSUN2 and SOAT2, along with elevated m5C levels in HCC tissues. In vitro experiments uncovered that NSUN2 augmented the reprogramming of energy metabolism and repressed the activity and cytotoxicity of CD8+ T cells, contributing to immune evasion. In vivo studies further substantiated the role of NSUN2 in fostering immune evasion and tumor formation of HCC by modulating the m5C modification of SOAT2. CONCLUSIONS The findings highlight the critical role of NSUN2 in driving HCC progression through the regulation of m5C modification on SOAT2. These findings present potential molecular markers for HCC diagnosis and therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Jinhua Jiang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Feng Liu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Dan Cui
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Caixia Xu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Jiachang Chi
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Tinghua Yan
- The First Clinical Medical College of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Fang Guo
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| |
Collapse
|
16
|
Lu L, Wang L, Yang M, Wang H. Role of METTL16 in PPARγ methylation and osteogenic differentiation. Cell Death Dis 2025; 16:271. [PMID: 40210616 PMCID: PMC11986173 DOI: 10.1038/s41419-025-07527-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: 06/23/2024] [Revised: 02/11/2025] [Accepted: 03/12/2025] [Indexed: 04/12/2025]
Abstract
Osteoporosis, a prevalent bone disease, is characterized by the deterioration of bone tissue microstructure and imbalanced osteogenesis. The regulatory role of PPARγ m6A methylation mediated by METTL16 remains poorly elucidated. This study utilized advanced single-cell RNA sequencing (scRNA-seq) and Bulk RNA-seq techniques to explore how METTL16 influences the osteogenic differentiation of Bone Marrow-Derived Mesenchymal Stem Cells (BMSCs) and its implication in osteoporosis. The research revealed that METTL16 enhances the suppression of osteogenic differentiation in BMSCs, while PPARγ is associated with BMSC ferroptosis. Mechanistically, METTL16 facilitates the m6A modification of PPARγ transcription, thereby promoting ferroptosis in BMSCs and impeding their osteogenic differentiation. The in vivo animal experiments confirmed the pivotal role of the METTL16-PPARγ axis in osteoporosis development in mice. These findings suggest that the regulation of PPARγ m6A methylation by METTL16, leading to ferroptosis, is a critical mechanism impacting BMSC osteogenic differentiation and the pathogenesis of osteoporosis.
Collapse
Affiliation(s)
- Liangjie Lu
- Department of Orthopedics, Ningbo Medical Center Li Huili Hospital, Li Huili Hospital Affiliated to Ningbo University, Ningbo, China.
| | - Lijun Wang
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Minjie Yang
- Department of Orthopaedics, Jiu jiang NO.1 People's Hospital, Jiu jiang, China
| | - Huihan Wang
- Department of Orthopaedics, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
17
|
Chen G, Liu J, Guo Y, Sun P. Mechanisms for Regulatory Effects of Exercise on Metabolic Diseases from the Lactate-Lactylation Perspective. Int J Mol Sci 2025; 26:3469. [PMID: 40331975 PMCID: PMC12027343 DOI: 10.3390/ijms26083469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 05/08/2025] Open
Abstract
Metabolic diseases, including cardiovascular diseases, type 2 diabetes mellitus (T2DM), osteoporosis, and non-alcoholic fatty liver disease (NAFLD), constitute a major global health burden associated with chronic morbidity and mortality. Lactate, once considered as a metabolic byproduct, has emerged as a key regulator of cellular reprogramming through lactylation, a novel post-translational modification (PTM) that dynamically couples metabolic flux to chromatin remodeling. Lactylation exerts dual regulatory roles as a signaling molecule via GPR81/GPR4-mediated pathways and as a substrate for the covalent modification of histones and metabolic enzymes. Pathologically, chronic hyperlactatemia suppresses mitochondrial biogenesis, driving metabolic cardiomyopathy through the epigenetic silencing of oxidative metabolism genes. Conversely, exercise-induced lactate surges transiently enhance insulin sensitivity via AMPK/PGC-1α/GLUT4 signaling, resolve inflammation through GPR81-mediated M2 macrophage polarization, and restore mitochondrial function via lactylation-dependent pathways. This review delineates lactylation as a spatiotemporal rheostat: chronic dysregulation perpetuates metabolic disorders, whereas acute exercise-mediated lactylation remodels transcriptional networks to restore metabolic homeostasis. Future research should integrate multiomics to clarify lactylation's spatiotemporal dynamics, tissue-specific thresholds, metabolism-immunity interactions, and metabolic-epigenetic crosstalk for the precision management of metabolic diseases.
Collapse
Affiliation(s)
- Guannan Chen
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; (G.C.); (J.L.); (Y.G.)
| | - Jinchao Liu
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; (G.C.); (J.L.); (Y.G.)
| | - Yilan Guo
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; (G.C.); (J.L.); (Y.G.)
| | - Peng Sun
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; (G.C.); (J.L.); (Y.G.)
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention of the Ministry of Education, East China Normal University, Shanghai 200241, China
| |
Collapse
|
18
|
Kwas K, Szubert M, Wilczyński JR. Latest Update on lncRNA in Epithelial Ovarian Cancer-A Scoping Review. Cells 2025; 14:555. [PMID: 40214508 PMCID: PMC11988607 DOI: 10.3390/cells14070555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) are RNA molecules exceeding 200 nucleotides that do not encode proteins yet play critical roles in regulating gene expression at multiple levels, such as chromatin modification and transcription. These molecules are significantly engaged in cancer progression, development, metastasis, and chemoresistance. However, the function of lncRNAs in epithelial ovarian cancer (EOC) has not yet been thoroughly studied. EOC remains challenging due to its complex molecular pathogenesis, characterized by genetic and epigenetic alterations. Emerging evidence suggests that lncRNAs, such as XIST, H19, NEAT1, and MALAT1, are involved in EOC by modulating gene expression and signaling pathways, influencing processes like cell proliferation, invasion, migration, and chemoresistance. Despite extensive research, the precise mechanism of acting of lncRNAs in EOC pathogenesis and treatment resistance still needs to be fully understood, highlighting the need for further studies. This review aims to provide an updated overview of the current understanding of lncRNAs in EOC, emphasizing their potential as biomarkers and therapeutic targets. We point out the gaps in the knowledge regarding lncRNAs' influence on epithelial ovarian cancer (EOC), deliberating on new possible research areas.
Collapse
Affiliation(s)
- Katarzyna Kwas
- Department of Surgical and Oncologic Gynaecology, 1st Department of Gynaecology and Obstetrics, Medical University of Lodz, 90-136 Łódź, Poland; (M.S.); (J.R.W.)
| | | | | |
Collapse
|
19
|
Hooshangnejad H, Lee J, Bell L, Hales RK, Voong KR, Han‐Oh S, Ding K, Farjam R. Quantitative beam optimization for radiotherapy of peripheral lung lesions: A pilot study in stereotactic body radiotherapy. J Appl Clin Med Phys 2025; 26:e70029. [PMID: 39985560 PMCID: PMC11969078 DOI: 10.1002/acm2.70029] [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: 09/13/2024] [Revised: 12/16/2024] [Accepted: 01/01/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND To quantify beam optimization for stereotactic body radiotherapy (SBRT) of peripheral lung lesions. METHOD The new beam optimization approach was based on maximizing the therapeutic gain (TG) of the beam set by minimizing the average physical depth of the lesion with respect to the beam's eye view (BEV). The new approach was evaluated by replanning the 25 SBRT lesions retrospectively to assess if a better plan is achievable in all aspects. Difference in 25 Gy isodose line volume (IDLV25 Gy), IDLV20 Gy, IDLV15 Gy, IDLV10 Gy, and IDLV5 Gy between the two plan cohorts were calculated as a measure of plan size and fitted in a linear regression model against the changes in the lesion depth with respect to the BEV to assess the relationship between the changes in the treatment depth and that of the plan size. RESULTS Beam optimization achieved a better plan in all cases by lowering the depth of treatment with an average of % 20.03 ± 12.30 (3.66%-45.78%). As the depth of treatment decreases, the size of the plan also decreases. We observed a reduction of % 4.64 ± 4.55 (0.02%-21.58%, p < 3.8 × 10-5), %5.16 ± 5.54 (0.03%-24.68%, p < 0.005), %6.46 ± 6.95 (-1.35%-29.05%, p < 0.009), %12.83 ± 9.06 (0.89%-37.65%, p < 0.0001), and %14.01 ± 9.87 (1.43%-41.84%, p < 4.5 × 10-6) in IDLV25 Gy, IDLV20 Gy, IDLV15 Gy, IDLV10 Gy, and IDLV5 Gy, respectively. CONCLUSION Physical depth of the lesion with respect to the BEV is inversely proportional to the TG of a beam-set and can be used as a robust and standard metric to select an appropriate beam-set for SBRT of the peripheral lung lesions. Further evaluation warrants the utility of such concept in routine clinical use.
Collapse
Affiliation(s)
- Hamed Hooshangnejad
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
- Carnegie Center for Surgical InnovationJohns Hopkins UniversityBaltimoreMarylandUSA
- Department of Radiation Oncology and Molecular SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jina Lee
- Department of Radiation Oncology and Molecular SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Leslie Bell
- Department of Radiation Oncology and Molecular SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Russell K. Hales
- Department of Radiation Oncology and Molecular SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Khinh Ranh Voong
- Department of Radiation Oncology and Molecular SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Sarah Han‐Oh
- Department of Radiation Oncology and Molecular SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Kai Ding
- Carnegie Center for Surgical InnovationJohns Hopkins UniversityBaltimoreMarylandUSA
- Department of Radiation Oncology and Molecular SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Reza Farjam
- Department of Radiation Oncology and Molecular SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
20
|
Marsh SGE. Nomenclature for Factors of the HLA System, Update October, November and December 2024. Int J Immunogenet 2025; 52:88-123. [PMID: 39875331 PMCID: PMC11895856 DOI: 10.1111/iji.12707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 01/17/2025] [Indexed: 01/30/2025]
|
21
|
Li Y, Piao Z, Ge X, Feng J, Sun D, Zhang J. Environmental pollutants and rectal cancer: The impact of water contamination. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 294:118072. [PMID: 40127547 DOI: 10.1016/j.ecoenv.2025.118072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/14/2025] [Accepted: 03/14/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND Water is a fundamental resource for life, and exposure to water contamination has far-reaching implications for an increased risk of tumor diseases. METHODS Studies of rectal and colorectal cancer related to water contamination were identified from the published literature in the PUBMED databases from 2010 to 2024. RESULTS This review provides a critical analysis of the current evidence, summarizing the association of water contamination, including industrial waste, pesticides, heavy metals, with rectal and colorectal cancer. It highlights their impact on rectal and colorectal cancer progression by underlying processes of DNA damage, chronic inflammation, and microbial contamination. CONCLUSION Rectal cancer is a significant global health concern with a strong association between environmental pollutants in water sources and increased incidence of rectal cancer. It is vital to identify how waster pollutants influence the development and progression of rectal cancer and formulate targeted preventive approaches and social interventions to decrease the disease's impact.
Collapse
Affiliation(s)
- Yezhou Li
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Zhe Piao
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xinbin Ge
- Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Jinbao Feng
- Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Denghua Sun
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China.
| | - Jiayu Zhang
- Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China.
| |
Collapse
|
22
|
Aparo A, Avesani S, Parmigiani L, Napoli S, Bertoni F, Bonnici V, Cascione L, Giugno R. EasyCircR: Detection and reconstruction of circular RNAs post-transcriptional regulatory interaction networks. Comput Biol Med 2025; 188:109846. [PMID: 39987699 DOI: 10.1016/j.compbiomed.2025.109846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 02/25/2025]
Abstract
Circular RNAs (circRNAs) are regulatory RNAs that play a crucial role in various biological activities and have been identified as potential biomarkers for neurological disorders and cancer. CircRNAs have emerged as significant regulators of gene expression through different mechanisms, including regulation of transcription and splicing, modulation of translation, and post-translational modifications. Additionally, some circRNAs operate as microRNA (miRNA) sponges in the cytoplasm, boosting post-transcriptional expression of target genes by inhibiting miRNA activity. Although existing pipelines can reconstruct circRNAs, identify miRNAs sponged by them, retrieve cascade-regulated mRNAs, and represent the regulatory interactions as complex circRNA-miRNA-mRNA networks, none of the state-of-the-art approaches can discriminate the biological level at which the mRNAs involved in the interactions are regulated, avoiding considering potential target mRNAs not regulated at the post-transcriptional level. EasyCircR is a novel R package that combines circRNA detection and reconstruction with post-transcriptional gene expression analysis (exon-intron split analysis) and miRNA response element prediction. The package enables estimation and visualization of circRNA-miRNA-mRNA interactions through an intuitive Shiny application, leveraging the post-transcriptional regulatory nature of circRNA-miRNA relationship and excluding unrealistic regulatory interactions at the biological level. EasyCircR source code, Docker container and user guide are available at: https://github.com/InfOmics/EasyCircR.
Collapse
Affiliation(s)
- Antonino Aparo
- Department of Computer Science, University of Verona, Strada le Grazie, 15, Verona, 37134, Italy; Research Center LURM (Interdepartmental Laboratory of Medical Research), University of Verona, Verona, 37134, Italy
| | - Simone Avesani
- Department of Computer Science, University of Verona, Strada le Grazie, 15, Verona, 37134, Italy.
| | - Luca Parmigiani
- Faculty of Technology and Center for Biotechnology (CeBiTec), Bielefeld, 33615, Germany; Bielefeld Institute for Bioinformatics Infrastructure (BIBI), Bielefeld, 33615, Germany; Graduate School "Digital Infrastructure for the Life Sciences"(DILS), Bielefeld, 33615, Germany
| | - Sara Napoli
- Institute of Oncology Research, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Bellinzona, 6500, Switzerland
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Bellinzona, 6500, Switzerland; Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, 6500, Switzerland
| | - Vincenzo Bonnici
- Department of Mathematical, Physical and Computer Sciences, University of Parma, Parma, 43124, Italy
| | - Luciano Cascione
- Institute of Oncology Research, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Bellinzona, 6500, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, 1015, Switzerland
| | - Rosalba Giugno
- Department of Computer Science, University of Verona, Strada le Grazie, 15, Verona, 37134, Italy
| |
Collapse
|
23
|
Ożarowski M, Karpiński TM, Czerny B, Kamiński A, Seremak-Mrozikiewicz A. Plant Alkaloids as Promising Anticancer Compounds with Blood-Brain Barrier Penetration in the Treatment of Glioblastoma: In Vitro and In Vivo Models. Molecules 2025; 30:1561. [PMID: 40286187 PMCID: PMC11990316 DOI: 10.3390/molecules30071561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/08/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Glioblastoma (GBM) is one of the most invasive central nervous system tumors, with rising global incidence. Therapy resistance and poor prognosis highlight the urgent need for new anticancer drugs. Plant alkaloids, a largely unexplored yet promising class of compounds, have previously contributed to oncology treatments. While past reviews provided selective insights, this review aims to collectively compare data from the last decade on (1) plant alkaloid-based anticancer drugs, (2) alkaloid transport across the blood-brain barrier (BBB) in vitro and in vivo, (3) alkaloid mechanisms of action in glioblastoma models (in vitro, in vivo, ex vivo, and in silico), and (4) cytotoxicity and safety profiles. Additionally, innovative drug delivery systems (e.g., nanoparticles and liposomes) are discussed. Focusing on preclinical studies of single plant alkaloids, this review includes 22 botanical families and 28 alkaloids that demonstrated anti-GBM activity. Most alkaloids act in a concentration-dependent manner by (1) reducing glioma cell viability, (2) suppressing proliferation, (3) inhibiting migration and invasion, (4) inducing cell death, (5) downregulating Bcl-2 and key signaling pathways, (6) exhibiting antiangiogenic effects, (7) reducing tumor weight, and (8) improving survival rates. The toxic and adverse effect analysis suggests that alkaloids such as noscapine, lycorine, capsaicin, chelerythrine, caffeine, boldine, and colchicine show favorable therapeutic potential. However, tetrandrine, nitidine, harmine, harmaline, cyclopamine, cocaine, and brucine may pose greater risks than benefits. Piperine's toxicity and berberine's poor bioavailability suggest the need for novel drug formulations. Several alkaloids (kukoamine A, cyclovirobuxine D, α-solanine, oxymatrine, rutaecarpine, and evodiamine) require further pharmacological and toxicological evaluation. Overall, while plant alkaloids show promise in glioblastoma therapy, progress in assessing their BBB penetration remains limited. More comprehensive studies integrating glioma research and advanced drug delivery technologies are needed.
Collapse
Affiliation(s)
- Marcin Ożarowski
- Department of Biotechnology, Institute of Natural Fibres and Medicinal Plants—National Research Institute, Wojska Polskiego 71b, 60-630 Poznań, Poland
| | - Tomasz M. Karpiński
- Chair and Department of Medical Microbiology, Poznań University of Medical Sciences, Rokietnicka 10, 60-806 Poznań, Poland
| | - Bogusław Czerny
- Department of General Pharmacology and Pharmacoeconomics, Pomeranian Medical University in Szczecin, Żołnierska 48, 70-204 Szczecin, Poland;
- Institute of Natural Fibres and Medicinal Plants—National Research Institute, Wojska Polskiego 71b, 60-630 Poznań, Poland
| | - Adam Kamiński
- Department of Orthopaedics and Traumatology, Independent Public Clinical Hospital No. 1, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland;
| | - Agnieszka Seremak-Mrozikiewicz
- Division of Perinatology and Women’s Disease, Poznań University of Medical Sciences, Polna 33, 60-535 Poznań, Poland;
- Laboratory of Molecular Biology in Division of Perinatology and Women’s Diseases, University of Medical Sciences, Polna 33, 60-535 Poznań, Poland
| |
Collapse
|
24
|
Cao Q, Sun D, Tu C, Wang J, Fu R, Gong R, Xiao Y, Liu Q, Li X. Defining gastric cancer ecology: the crucial roles of TREM2 + macrophages and fibroblasts in tumor microenvironments. Commun Biol 2025; 8:514. [PMID: 40155473 PMCID: PMC11953254 DOI: 10.1038/s42003-025-07512-2] [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: 06/11/2024] [Accepted: 01/10/2025] [Indexed: 04/01/2025] Open
Abstract
Gastric cancer (GC) remains a major global health challenge, characterized by a complex tumor microenvironment (TME) that significantly influences disease progression and therapeutic outcomes. This study focuses on TREM2+ lipid-associated macrophages (LAM) and cancer-associated fibroblasts (CAFs) in modulating the GC microenvironment. Utilizing advanced single-cell RNA sequencing and bulk RNA analyses, we elucidated the interactive mechanisms through which CAFs enhance the immunosuppressive capabilities of TREM2+ LAMs via the CXCL12-CXCR4 signaling axis. Our findings reveal that this interaction facilitates tumor proliferation and inhibits apoptotic processes in GC cells. In vitro experiments confirmed the modulation of this pathway significantly affects tumor cell viability and invasiveness, underscoring the critical roles of these cellular interactions in promoting GC progression. These insights present TREM2+ LAMs and CAFs as potential therapeutic targets, offering new avenues for improving outcomes in GC treatment.
Collapse
Affiliation(s)
- Qianqian Cao
- Cancer Center, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Dianshui Sun
- Cancer Center, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Can Tu
- Vascular Intervention Department, The First Affiliated Hospital of Ningbo University, Ningbo, PR China
| | - Jihua Wang
- Cancer Center, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Runjia Fu
- Cancer Center, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Rumei Gong
- Cancer Center, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Yueying Xiao
- Department of Spine Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Qin Liu
- Cancer Center, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Xiaomei Li
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China.
| |
Collapse
|
25
|
Wijetunga NA, Yahalom J, Imber BS. The art of war: using genetic insights to understand and harness radiation sensitivity in hematologic malignancies. Front Oncol 2025; 14:1478078. [PMID: 40191738 PMCID: PMC11968681 DOI: 10.3389/fonc.2024.1478078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/20/2024] [Indexed: 04/09/2025] Open
Abstract
It is well established that hematologic malignancies are often considerably radiosensitive, which enables usage of far lower doses of therapeutic radiotherapy. This review summarizes the currently known genomic landscape of hematologic malignancies, particularly as it relates to radiosensitivity and the field of radiation oncology. By tracing the historical development of the modern understanding of radiosensitivity, we focus on the discovery and implications of pivotal mutated genes in hematologic malignancies such as TP53, ATM, and other genes critical to DNA repair pathways. These genetic insights have contributed significantly to the advancement of personalized medicine, aiming to enhance treatment precision and outcomes, and there is an opportunity to extend these insights to personalized radiotherapy. We explore the transition from early discoveries to the current efforts in integrating comprehensive genomic data into clinical practice. Specific examples from Hodgkin lymphoma, non-Hodgkin lymphoma, and plasma cell neoplasms illustrate how genetic mutations could influence radiosensitivity and impact subsequent radiotherapeutic response. Despite the advancements, challenges remain in translating these genetic insights into routine clinical practice, particularly due to the heterogeneity of alterations and the complex interactions within cancer signaling pathways. We emphasize the potential of radiogenomics to address these challenges by identifying genetic markers that predict radiotherapy response and toxicity, thereby refining treatment strategies. The need for robust decision support systems, standardized protocols, and ongoing education for healthcare providers is critical to the successful integration of genomic data into radiation therapy. As research continues to validate genetic markers and explore novel therapeutic combinations, the promise of personalized radiotherapy becomes increasingly attainable, offering the potential to significantly improve outcomes for patients with hematologic malignancies.
Collapse
Affiliation(s)
- N. Ari Wijetunga
- Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, United States
| | - Joachim Yahalom
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Brandon S. Imber
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
26
|
Yan X, Niu Y, Yang X, Zhao R, Cui W, Guo X, Zhang J, Ma M. FDP/FIB ratio serves as a novel biomarker for diagnosing bone marrow invasion in gastric cancer and predicting patient prognosis\. Sci Rep 2025; 15:9462. [PMID: 40108276 PMCID: PMC11923145 DOI: 10.1038/s41598-025-93056-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
Objective This study aimed to identify laboratory indicators with significant implications for bone marrow invasion in gastric cancer patients and to evaluate their prognostic value. Methods A retrospective analysis of the clinical data of 320 gastric cancer patients who underwent either bone marrow cytological examination or bone marrow biopsy at our hospital between January 2013 and December 2023 was conducted. Among these patients, 31 patients with confirmed bone marrow invasion composed the study group, whereas 34 stage IV gastric cancer patients without bone marrow invasion composed the control group. Differences in demographic and laboratory data between the two groups were compared. Receiver operating characteristic curves were used to identify valuable indicators for predicting bone marrow invasion in patients with gastric cancer. Survival analysis was performed using the Kaplan‒Meier method and included the plotting of survival curves. Additionally, Cox proportional hazards regression analysis was performed to evaluate independent prognostic factors. Results Significantly different values (all P < 0.05) were observed for age, peripheral blood immature cells, Hb, PLT, SII, FIB, PT, FDP, D-Dimer, FDP/FIB, CEA, and CA72-4 between stage IV gastric cancer patients with and without bone marrow infiltration. The ROC analysis indicated that at a threshold value of 5.197 for FDP/FIB, the AUC was 0.958 (P < 0.01). Within the cohort of 65 stage IV gastric cancer patients, those with bone marrow invasion and high FDP/FIB ratios exhibited notably shorter median survival times than those without bone marrow invasion and with low FDP/FIB ratios (χ2 = 25.928, 20.128, P < 0.001). Multivariate analysis demonstrated that bone marrow invasion (HR = 4.148, P = 0.020) and the FDP/FIB ratio (HR = 1.026, P = 0.024) were independent risk factors influencing the prognosis and survival outcome of stage IV gastric cancer patients. Among the subset of 31 gastric cancer patients with bone marrow invasion, the median survival time for the high FDP/FIB group was 22 days, which was significantly shorter than the 60 days observed in the low FDP/FIB group (χ2 = 8.479, P = 0.004). Conclusion The FDP/FIB ratio can serve as an important indicator for the diagnosis and prognostic evaluation of bone marrow invasion in patients with gastric cancer.
Collapse
Affiliation(s)
- Xi Yan
- Department of Clinical Laboratory, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yinghao Niu
- Department of Clinical Biobank, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xingxiao Yang
- Department of Hospital Infection Management, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Riyang Zhao
- Department of Clinical Laboratory, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wenxuan Cui
- Department of Clinical Laboratory, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiujuan Guo
- Department of Clinical Laboratory, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China.
- , 12# Jiankang Road, Shijiazhuang, China.
| | - Jinyan Zhang
- Department of Clinical Laboratory, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China.
- , 12# Jiankang Road, Shijiazhuang, China.
| | - Ming Ma
- Department of Clinical Laboratory, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China.
- , 12# Jiankang Road, Shijiazhuang, China.
| |
Collapse
|
27
|
Lin YK, Zhu LL, Zhao J, Xiang ZL. Radiotherapy volume delineation based on MRI and 18F-FDG-PET/MRI in locally recurrent rectal cancer. Abdom Radiol (NY) 2025:10.1007/s00261-025-04859-2. [PMID: 40095014 DOI: 10.1007/s00261-025-04859-2] [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: 11/25/2024] [Revised: 02/19/2025] [Accepted: 02/23/2025] [Indexed: 03/19/2025]
Abstract
OBJECTIVE To evaluate the value of 18F-FDG-positron emission tomography (PET)/magnetic resonance imaging (MRI) functional imaging in the radiotherapy of locally recurrent rectal cancer by comparing the target volume delineation based on PET/MRI and MRI. MATERIALS AND METHODS Twenty-six patients who were diagnosed with locally recurrent rectal cancer were included in this study. Patients underwent PET/MRI, and the target volume was delineated independently by three radiation oncologists. The degree of overlap, spatial consistency, and difference in the target volume delineated based on the two methods were compared. The efficacy of PET/MRI and MRI in detecting metastatic lymph nodes was analyzed. RESULTS In radiotherapy for patients with recurrent rectal cancer, the gross tumor volume (GTV), clinical target area (CTV), and nodal gross tumor volume (GTVn) delineated based on MRI and PET/MRI were correlated (P < 0.001, P < 0.001, and P < 0.001, respectively). Differences in CTV were statistically significant (P < 0.001), and the CTV greatly overlapped spatially. There is spatial heterogeneity in GTV and GTVn based on the two imaging modalities. Metastatic lymph node analysis revealed that the detection efficiency of the two modalities was the same at the population level. There was no significant difference in the number of metastatic lymph nodes detected (P = 0.521). CONCLUSION PET/MRI can improve the accuracy of target volume delineation and has similar advantages to MRI in assessing the number of metastatic lymph nodes in patients with recurrent rectal cancer.
Collapse
Affiliation(s)
- Yu-Kun Lin
- Department of Radiation Oncology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai, China
| | - Lei-Lei Zhu
- Department of Radiation Oncology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai, China
| | - Jun Zhao
- Department of Nuclear Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai, China.
| | - Zuo-Lin Xiang
- Department of Radiation Oncology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai, China.
| |
Collapse
|
28
|
Jin YP, Li GW, Xu QQ, Wang XL. ZEB1 promotes the immune escape of ovarian cancer through the MCSF-CCL18 axis. Cancer Cell Int 2025; 25:95. [PMID: 40089803 PMCID: PMC11909986 DOI: 10.1186/s12935-025-03724-y] [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: 04/17/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
This study aimed to determine the molecular mechanisms underlying immune escape in ovarian cancer. Samples of ovarian cancer were used to explore the regulatory pathways involved in the malignant phenotype. Tumor cell models with different levels of factor expression were constructed via transfection, and their regulation was determined through investigation of protein expressions. Moreover, our study aimed to investigate the effects of M2 polarization and TAMs aggregation on the apoptosis of CD8 + T-cells, and determine their regulatory axis. Results revealed ZEB1 may promote CCL18 expression via upregulation of MCSF concentration. Notably, high CCL18 expression levels were associated with the aggregation of M2-TAMs and the apoptosis of CD8 + T-cells. In addition, results of the present study demonstrated that the proliferation and invasion of ovarian cancer cells with high expression levels of proteins associated with ZEB1 signal pathway were increased. At the same time the growth rate of tumors in mice was reduced following ZEB1 knockdown, and the volume/weight of tumors were markedly decreased both in vitro and in vivo. Moreover, our results revealed that the aggregation of M2-TAMs and the apoptosis of CD8 + T-cells were significantly decreased in tumor cells following ZEB1 knockdown. Thus, these results verified that ZEB1 may promote the M2 polarization of TAMs via the MCSF axis, leading to the increased secretion of CCL18. Moreover, the MCSF axis may mediate immune escape through the induction of CD8 + T-cell apoptosis, ultimately promoting the malignant phenotype in ovarian cancer cells.
Collapse
Affiliation(s)
- Yan-Ping Jin
- School of Nursing, Jiangsu Health Vocational College, Nanjing, 211899, Jiangsu, China
| | - Guo-Wei Li
- School of Rehabilitation Science, Nanjing Normal University of Special Education, Nanjing, 210038, Jiangsu, China.
| | - Qian-Qian Xu
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital North), Suzhou, 215008, China
| | - Xiao-Lan Wang
- Department of Obstetrics and Gynecology, Zhongda Hospital Jiangbei Branch, School of Medicine, Southeast University, Nanjing, 210048, Jiangsu, China
| |
Collapse
|
29
|
Lin QN, Wu Q, Lv C, Tang J, Zhao LY, Xu KY. Potential role of coagulation markers in early detection of bone metastasis in gastric cancer: A critical review. World J Gastrointest Oncol 2025; 17:100292. [PMID: 40092945 PMCID: PMC11866228 DOI: 10.4251/wjgo.v17.i3.100292] [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: 08/13/2024] [Revised: 11/26/2024] [Accepted: 12/17/2024] [Indexed: 02/14/2025] Open
Abstract
This letter to the editor involves the article published in the World Journal of Gastrointestinal Oncology. Bone metastasis (BM) in gastric cancer (GC) is uncommon but can be prevalent in autopsy studies. BM significantly impairs quality of life and is frequently underdiagnosed, as sensitive diagnostic tests are only performed after symptoms occur. Imaging is crucial for the diagnosis of BM but is not routinely used for screening and is expensive. Examining laboratory risk factors for BM in GC patients using multivariate analysis could be a more effective approach.
Collapse
Affiliation(s)
- Qing-Nan Lin
- School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
- Department of Orthopedics, Wuyi County Traditional Chinese Medicine Hospital, Jinhua 321200, Zhejiang Province, China
| | - Qian Wu
- Operating Room, Wuyi County Traditional Chinese Medicine Hospital, Jinhua 321200, Zhejiang Province, China
| | - Chong Lv
- Department of Orthopedics, Wuyi County Traditional Chinese Medicine Hospital, Jinhua 321200, Zhejiang Province, China
| | - Jun Tang
- Department of Orthopedics, Wuyi County Traditional Chinese Medicine Hospital, Jinhua 321200, Zhejiang Province, China
| | - Ling-Yun Zhao
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Ke-Yang Xu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China
| |
Collapse
|
30
|
Mu P, Mo S, He X, Zhang H, Lv T, Xu R, He L, Xia F, Zhou S, Chen Y, Wang Y, Shen L, Wan J, Huang L, Lu W, Liang X, Li X, Lu P, Peng J, Hua G, Hu K, Zhang Z, Wang Y. Unveiling radiobiological traits and therapeutic responses of BRAF V600E-mutant colorectal cancer via patient-derived organoids. J Exp Clin Cancer Res 2025; 44:92. [PMID: 40069844 PMCID: PMC11895145 DOI: 10.1186/s13046-025-03349-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/21/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Radiotherapy (RT) is an essential treatment for colorectal cancer (CRC), yet the factors influencing radiosensitivity remain unclear. In the quest to enhance the therapeutic efficacy in CRC, the interplay between genetic mutations and RT sensitivity has emerged as a pivotal yet enigmatic area. METHODS We harness the fidelity of patient-derived organoids (PDOs) to dissect the molecular landscape of radiosensitivity, with a particular emphasis on BRAFV600E mutations. To further investigate, a cohort of 9 BRAFV600E-mutant and 10 BRAF wild-type PDOs is constructed to systematically assess the radiobiological traits of BRAFV600E-mutant CRC, including morphology, cell viability, and DNA damage, while also evaluating their responses to chemotherapy and chemoradiotherapy. RESULTS Our systematic investigation unveils a profound correlation between BRAFV600E mutation status and radioresistance, which is validated by clinical treatment responses. Intriguingly, BRAFV600E-mutant PDOs exhibit reduced sensitivity to conventional chemotherapy, yet demonstrate an enhanced response to combined chemoradiotherapy, characterized by increased apoptosis. The results are validated through in vivo analyses using patient-derived organoid xenograft mouse models and aligned with patient clinical outcomes. CONCLUSIONS This study outlines the distinct radiobiological profile of BRAFV600E-mutant CRC, underscoring the critical role of radiotherapy in comprehensive treatment strategies. This work not only advances our molecular understanding of CRC but also paves the way for precision medicine, offering valuable insights for therapeutic decision-making in the clinical management of BRAFV600E-mutant CRC.
Collapse
Affiliation(s)
- Peiyuan Mu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Shaobo Mo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Xingfeng He
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Hui Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Tao Lv
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Ruone Xu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Luoxi He
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Fan Xia
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Shujuan Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Yajie Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Yaqi Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Lijun Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Juefeng Wan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Lili Huang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Weiqing Lu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Xinyue Liang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
- Cancer institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Xiaomeng Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Urology Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Ping Lu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Junjie Peng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Guoqiang Hua
- D1Med Technology (Shanghai) Inc, Shanghai, 201802, China
| | - Kewen Hu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China.
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China.
- Cancer institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China.
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China.
| | - Yan Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China.
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China.
| |
Collapse
|
31
|
Wang Y, Pan Z, Cai H, Li S, Huang Y, Zhuang J, Liu X, Guan G. Prognostic model for log odds of negative lymph node in locally advanced rectal cancer via interpretable machine learning. Sci Rep 2025; 15:7924. [PMID: 40050297 PMCID: PMC11885450 DOI: 10.1038/s41598-025-90191-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 02/11/2025] [Indexed: 03/09/2025] Open
Abstract
No studies have examined the prognostic value of the log odds of negative lymph nodes/T stage (LONT) in locally advanced rectal cancer (LARC) treated with neoadjuvant chemoradiotherapy (nCRT). We aimed to assess the prognostic value of LONT and develop a machine learning model to predict overall survival (OS) and disease-free survival (DFS) in LARC patients treated with nCRT. The study included 820 LARC patients who received nCRT between September 2010 and October 2017. Univariate and multivariate Cox regression analyses identified prognostic factors, which were then used to develop risk assessment models with 9 machine learning algorithms. Model hyperparameters were optimized using random search and 10-fold cross-validation. The models were evaluated using metrics such as the area under the receiver operating characteristic curves (AUC), decision curve analysis, calibration curves, and precision and accuracy for predicting OS and DFS. Shapley's additive explanations (SHAP) was also used for model interpretation. The study included 820 patients, identifying LONT as a significant independent prognostic factor for both OS and DFS. Nine machine learning algorithms were used to create predictive models based on these factors. The extreme gradient boosting (XGB) model showed the best performance, with a mean AUC of 0.89 for OS and 0.83 for DFS in 10-fold cross-validation. Additionally, the predictions generated by the XGB model were analyzed using SHAP. Finally, we developed an online web-based calculator utilizing the XGB model to enhance the model's generalizability and to provide improved support for physicians in their decision-making processes. The study developed an XGB model utilizing LONT to predict OS and DFS in patients with LARC undergoing nCRT. Furthermore, an online web calculator was constructed using the XGB model to facilitate the model's generalization and to enhance physician decision-making.
Collapse
Affiliation(s)
- Ye Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zhen Pan
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Huajun Cai
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Shoufeng Li
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ying Huang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jinfu Zhuang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xing Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guoxian Guan
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- Department of Colorectal Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fuzhou, China.
| |
Collapse
|
32
|
Nie T, Nepovimova E, Wu Q. Circadian rhythm, hypoxia, and cellular senescence: From molecular mechanisms to targeted strategies. Eur J Pharmacol 2025; 990:177290. [PMID: 39863143 DOI: 10.1016/j.ejphar.2025.177290] [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: 10/25/2024] [Revised: 01/03/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Cellular senescence precipitates a decline in physiological activities and metabolic functions, often accompanied by heightened inflammatory responses, diminished immune function, and impaired tissue and organ performance. Despite extensive research, the mechanisms underpinning cellular senescence remain incompletely elucidated. Emerging evidence implicates circadian rhythm and hypoxia as pivotal factors in cellular senescence. Circadian proteins are central to the molecular mechanism governing circadian rhythm, which regulates homeostasis throughout the body. These proteins mediate responses to hypoxic stress and influence the progression of cellular senescence, with protein Brain and muscle arnt-like 1 (BMAL1 or Arntl) playing a prominent role. Hypoxia-inducible factor-1α (HIF-1α), a key regulator of oxygen homeostasis within the cellular microenvironment, orchestrates the transcription of genes involved in various physiological processes. HIF-1α not only impacts normal circadian rhythm functions but also can induce or inhibit cellular senescence. Notably, HIF-1α may aberrantly interact with BMAL1, forming the HIF-1α-BMAL1 heterodimer, which can instigate multiple physiological dysfunctions. This heterodimer is hypothesized to modulate cellular senescence by affecting the molecular mechanism of circadian rhythm and hypoxia signaling pathways. In this review, we elucidate the intricate relationships among circadian rhythm, hypoxia, and cellular senescence. We synthesize diverse evidence to discuss their underlying mechanisms and identify novel therapeutic targets to address cellular senescence. Additionally, we discuss current challenges and suggest potential directions for future research. This work aims to deepen our understanding of the interplay between circadian rhythm, hypoxia, and cellular senescence, ultimately facilitating the development of therapeutic strategies for aging and related diseases.
Collapse
Affiliation(s)
- Tong Nie
- College of Life Science, Yangtze University, Jingzhou, 434025, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, 434025, China.
| |
Collapse
|
33
|
Jacome MA, Wu Q, Chen J, Mohamed ZS, Mokhtari S, Piña Y, Etame AB. Molecular Underpinnings of Brain Metastases. Int J Mol Sci 2025; 26:2307. [PMID: 40076927 PMCID: PMC11900073 DOI: 10.3390/ijms26052307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Brain metastases are the most commonly diagnosed type of central nervous system tumor, yet the mechanisms of their occurrence are still widely unknown. Lung cancer, breast cancer, and melanoma are the most common etiologies, but renal and colorectal cancers have also been described as metastasizing to the brain. Regardless of their origin, there are common mechanisms for progression to all types of brain metastases, such as the creation of a suitable tumor microenvironment in the brain, priming of tumor cells, adaptations to survive spreading in lymphatic and blood vessels, and development of mechanisms to penetrate the blood-brain barrier. However, there are complex genetic and molecular interactions that are specific to every type of primary tumor, making the understanding of the metastatic progression of tumors to the brain a challenging field of study. In this review, we aim to summarize current knowledge on the pathophysiology of brain metastases, from specific genetic characteristics of commonly metastatic tumors to the molecular and cellular mechanisms involved in progression to the central nervous system. We also briefly discuss current challenges in targeted therapies for brain metastases and how there is still a gap in knowledge that needs to be overcome to improve patient outcomes.
Collapse
Affiliation(s)
- Maria A. Jacome
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Qiong Wu
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (Q.W.); (J.C.); (S.M.); (Y.P.)
| | - Jianan Chen
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (Q.W.); (J.C.); (S.M.); (Y.P.)
| | | | - Sepideh Mokhtari
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (Q.W.); (J.C.); (S.M.); (Y.P.)
| | - Yolanda Piña
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (Q.W.); (J.C.); (S.M.); (Y.P.)
| | - Arnold B. Etame
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (Q.W.); (J.C.); (S.M.); (Y.P.)
| |
Collapse
|
34
|
Awang D, Danzeng K, Wang T, Deji Q, Huang M, Ren H, Liu X, Zhao B, Gongga L. CircRNA-miRNA-mRNA regulatory network in high-altitude hypobaric hypoxia-induced hearing impairment and hearing acclimatization. Braz J Otorhinolaryngol 2025; 91:101557. [PMID: 39874809 PMCID: PMC11808620 DOI: 10.1016/j.bjorl.2024.101557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 01/30/2025] Open
Abstract
OBJECTIVE High altitude hypobaric hypoxia can induce hearing impairment and hearing acclimatization, but few studies have been performed to decipher the potential transition between the two states. To decipher transition-related circular RNAs (circRNAs)-microRNAs (miRNAs)-messenger RNA (mRNAs) regulatory network. METHODS Wistar rats were airlifted from plain to high altitude and maintained for 30 days and 60 days. Hearing acclimatization was determined using the Auditory Brainstem Response (ABR) test. Cochlea tissues were isolated, and high-throughput circRNA analysis and mRNAs-sequencing were performed. Differentially Expressed circRNAs (DEcircRNAs) and Differentially Expressed mRNAs (DEmRNAs) were obtained, and circRNA-miRNA and miRNA-mRNA regulation were predicted. A circRNA-miRNA-mRNA competing endogenous RNA (ceRNA) network was also constructed. The DEmRNAs in this network were functionally annotated using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses based on Metascape. RESULTS The ABR assay indicated that hearing impairment happened on day 30 and hearing acclimatization occurred on day 60. Hearing impairment-related circRNAs (64 upregulated and 147 downregulated) and genes (572 upregulated and 757 downregulated) were identified. Hearing acclimatization-related circRNAs (79 upregulated and 142 downregulated) and genes (690 upregulated and 751 downregulated) were also identified. Hearing impairment and hearing acclimatization ceRNA networks were also constructed after integrating the predicted miRNA regulation analyses. Anterograde trans-synaptic signaling (GO:0098916) and negative regulation of cellular response to growth factor stimulus (GO:0090288) were regulated by hearing impairment ceRNA networks, and embryonic organ development (GO:0048568) was regulated by hearing acclimatization ceRNA networks. CONCLUSION Hearing impairment- and hearing acclimatization-associated circRNAs and ceRNA networks were identified, which contribute new knowledge to our understanding of acclimatization transition.
Collapse
Affiliation(s)
- Danzeng Awang
- Medical College, Tibet University, Department of Clinical Medicine, Lhasa, China; Tibet University, Medical College, Lhasa, China
| | - Kanzi Danzeng
- Lhasa People's Hospital, Children's Surgery Department, Lhasa, China
| | - Tianheng Wang
- Health Service Center of Jiri Street Office, Chengguan District, Lhasa, China
| | - Quzong Deji
- Tibet University, Medical College, Lhasa, China
| | - Mengting Huang
- Health Service Center of Jiri Street Office, Chengguan District, Lhasa, China
| | - Hailong Ren
- Tibet University, Medical College, Lhasa, China
| | - Xinzhu Liu
- Tibet University, Medical College, Lhasa, China
| | | | - Lanzi Gongga
- Tibet University, Medical College, Lhasa, China.
| |
Collapse
|
35
|
Shah C, Kruse M, Al-Hilli Z. Reimagining Deintensification for Low-Risk Breast Cancer. JCO Oncol Pract 2025; 21:323-332. [PMID: 39405491 DOI: 10.1200/op-24-00538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/01/2024] [Accepted: 09/18/2024] [Indexed: 03/15/2025] Open
Abstract
As outcomes for low-risk breast cancer continue to improve, research and clinical paradigms are increasingly focused on appropriate deintensification with the goal of improving the therapeutic ratio of breast cancer treatment. These deintensification approaches span across disciplines including breast surgery, radiation therapy, and systemic therapy. With regard to breast surgery, studies have continued to push deintensification when it comes to surgical margins with breast conservation, reducing re-excision rates, whereas deintensification of axillary surgery has reduced the rates of axillary lymph node dissection and increasingly the need for any axillary surgery, including sentinel lymph node biopsy for low-risk patients. With regard to radiation therapy, studies have allowed for a drastic reduction in treatment duration, whereas approaches that reduce the target of treatment have led to a change from from treatment daily for 5-7 weeks to many low-risk patients completing treatment in just five treatments. With regard to systemic therapy, use of genomic assays and tumor biology has led to reduced utilization of cytotoxic chemotherapy, with studies also allowing for dose reduction of endocrine therapy for patients with ductal carcinoma in situ. Moving forward, greater focus should be placed on interdisciplinary deintensification approaches such as the consideration of radiation therapy alone as compared with endocrine therapy alone for low-risk breast cancers.
Collapse
Affiliation(s)
- Chirag Shah
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Megan Kruse
- Department of Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Zahraa Al-Hilli
- Breast Center, Integrated Surgical Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
36
|
Golestanifar A, Khedri H, Noorabadi P, Saberiyan M. Identification of hub genes, non-coding RNAs and pathways in Renal cell carcinoma (RCC): A comprehensive in silico study. Biochem Biophys Rep 2025; 41:101942. [PMID: 39980583 PMCID: PMC11840516 DOI: 10.1016/j.bbrep.2025.101942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 02/22/2025] Open
Abstract
Backgrounds Renal cell carcinoma (RCC) is the most common type of kidney cancer in adults. RCC begins in the renal tubule epithelial cells, essential for blood filtration and urine production. Methods In this study, we aim to uncover the molecular mechanisms underlying kidney renal clear cell carcinoma (KIRC) by analyzing various non-coding RNAs (ncRNAs) and protein-coding genes involved in the disease. Using high-throughput sequencing datasets from the Gene Expression Omnibus (GEO), we identified differentially expressed mRNAs (DEMs), miRNAs (DEMIs), and circRNAs (DECs) in KIRC samples compared to normal kidney tissues. Our approach combined differential expression analysis, functional enrichment through Gene Ontology (GO) and KEGG pathway mapping, and a Protein-Protein Interaction (PPI) network to identify crucial hub genes in KIRC progression. Results Key findings include the identification of hub genes such as EGFR, FN1, IL6, and ITGAM, which were closely associated with immune responses, cell signaling, and metabolic dysregulation in KIRC. Further analysis indicated that these genes could be potential biomarkers for prognosis and therapeutic targets. We constructed a competitive endogenous RNA (ceRNA) network involving lncRNAs, circRNAs, and miRNAs, suggesting complex regulatory interactions that drive KIRC pathogenesis.Additionally, the study examined drug sensitivity associated with the expression of hub genes, revealing the potential for personalized treatments. Immune cell infiltration patterns showed significant correlations with hub gene expression, highlighting the importance of immune modulation in KIRC. Conclusion This research provides a foundation for developing targeted therapies and diagnostic biomarkers for KIRC while underscoring the need for experimental validation to confirm these bioinformatics insights.
Collapse
Affiliation(s)
- Ahmad Golestanifar
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Hengameh Khedri
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Parisa Noorabadi
- Department of Internal Medicine, School of Medicine, Urmia University of Medical sciences, Urmia, Iran
| | - Mohammadreza Saberiyan
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
37
|
Tatsuno S, Doi H, Inada M, Fukuda J, Ishida N, Uehara T, Nakamatsu K, Hosono M, Kawamura J, Matsuo Y. Intensity-modulated radiation therapy can reduce acute toxicities in long-course neoadjuvant radiation therapy combined with S-1 for locally advanced rectal cancer. Int J Clin Oncol 2025; 30:504-513. [PMID: 39812929 DOI: 10.1007/s10147-024-02690-1] [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: 09/04/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND The purpose of this study was to compare outcomes and adverse events between three-dimensional conformal radiation therapy (3D-CRT) and intensity-modulated radiation therapy (IMRT) in patients undergoing long-course neoadjuvant radiation therapy (NA-RT) for locally advanced rectal adenocarcinoma (LARC). METHODS We retrospectively analyzed a total of 47 consecutive patients who received NA-RT for LARC between January 2011 and September 2022. Seven and 40 patients were diagnosed with clinical stages II and III, respectively. The prescribed dose per fraction was 1.8 Gy for total doses of 45 or 50.4 Gy. Seventeen and 30 patients received 3D-CRT and IMRT, respectively. NA-RT was delivered with concurrent chemotherapy of oral administration of S-1. RESULTS Planned NA-RT was completed without any treatment interruption in 43 of the 47 patients. Two patients experienced treatment interruption, and two patients discontinued due to grade ≥ 3 toxicities. No significant differences were observed between patients receiving 3D-CRT and IMRT in local control, progression-free survival, and overall survival (P = 0.488, 0.259, and 0.636, respectively). Patients receiving IMRT showed significantly fewer non-hematological grade ≥ 2 acute toxicities than those receiving 3D-CRT (33.3% vs. 70.6%, P = 0.018). In addition, patients who received IMRT tended to have less intestinal toxicity of grade ≥ 2 than those who received 3D-CRT (P = 0.057). CONCLUSION IMRT significantly reduced grade ≥ 2 acute toxicities without compromising oncologic outcomes compared to 3D-CRT. Therefore, IMRT may be considered as a current standard treatment in the total neoadjuvant therapy era.
Collapse
Affiliation(s)
- Saori Tatsuno
- Department of Radiation Oncology, Kindai University Faculty of Medicine, 377-2, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Hiroshi Doi
- Department of Radiation Oncology, Kindai University Faculty of Medicine, 377-2, Ohno-Higashi, Osaka-Sayama, Osaka, Japan.
| | - Masahiro Inada
- Department of Radiation Oncology, Kindai University Faculty of Medicine, 377-2, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Junki Fukuda
- Department of Radiation Oncology, Kindai University Faculty of Medicine, 377-2, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Naoko Ishida
- Department of Radiation Oncology, Kindai University Faculty of Medicine, 377-2, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Takuya Uehara
- Department of Radiation Oncology, Kindai University Faculty of Medicine, 377-2, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Kiyoshi Nakamatsu
- Department of Radiation Oncology, Kindai University Faculty of Medicine, 377-2, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Makoto Hosono
- Department of Radiation Oncology, Kindai University Faculty of Medicine, 377-2, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Junichiro Kawamura
- Department of Surgery, Kindai University Faculty of Medicine, 377-2, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Yukinori Matsuo
- Department of Radiation Oncology, Kindai University Faculty of Medicine, 377-2, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| |
Collapse
|
38
|
Wang J, Cheng Y, Xiaoran Y, Chen F, Jie W, Yahui H, Yue W, Dong L, Yumei L, Cheng F, Libo Z, Jun Z. Globular adiponectin induces esophageal adenocarcinoma cell pyroptosis via the miR-378a-3p/UHRF1 axis. ENVIRONMENTAL TOXICOLOGY 2025; 40:429-444. [PMID: 38572808 DOI: 10.1002/tox.24266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/14/2024] [Accepted: 03/23/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Antiapoptosis is a major factor in the resistance of tumor cells to chemotherapy and radiotherapy. Thus, activation of cell pyroptosis may be an effective option to deal with antiapoptotic cancers such as esophageal adenocarcinoma (EAC). METHODS Differential expression of ubiquitin-like versus PHD and ring finger structural domain 1 (UHRF1) in EAC and near normal tissues was analyzed, as well as the prognostic impact on survival in EAC. Also, the same study was done for globular adiponectin (gAD). Simultaneously, the mRNA expression of UHRF1 was observed in different EAC cell lines. Real time cellular analysis (RTCA) was used to detect cell proliferation, and flow cytometry and inverted fluorescence microscopy were used to detect pyroptosis. Biocredit analysis was conducted to observe the correlation between UHRF1 and key pyroptosis proteins. OD values and CCK8 assay were used to determine the effect of miR-378a-3p on EAC cells. Quantitative real-time polymerase chain reaction and Western blot were used to detect the correlation between UHRF1, gAD, and miR-378a-3p in EAC cells. Moreover, in vivo and in vitro experiments were performed to detect the relevant effects on tumor migration and invasion after inhibiting UHRF1 expression. RESULTS UHRF1 was negatively correlated with the survival of patients with EAC, while miR-378a-3p showed the opposite effect. Additionally, gAD promoted EAC cell pyroptosis, upregulated miR-378a-3p, and significantly inhibited the proliferation of EAC cells. gAD directly reduced UHRF1 expression in EAC cells by upregulating miR-378a-3p. In cell migration and invasion assays, inhibition of UHRF1 expression significantly suppressed EAC cell metastasis. In animal experiments, we again demonstrated that gAD induced pyroptosis in EAC cells by inhibiting the expression of UHRF1. CONCLUSION gAD-induced upregulation of miR-378a-3p significantly inhibited the proliferation of EAC by targeting UHRF1. Therefore, gAD may serve as an alternative therapy for chemotherapy- and radiation-refractory EAC or other cancers with the same mechanism of pyroptosis action.
Collapse
Affiliation(s)
- Jun Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Department of Gastroenterology, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi, China
| | - Yan Cheng
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yin Xiaoran
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fengrong Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wu Jie
- Department of Pathology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Huang Yahui
- Department of Gastroenterology, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi, China
| | - Wang Yue
- Department of Gastroenterology, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi, China
| | - Liu Dong
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Luo Yumei
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Feng Cheng
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhang Libo
- Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhang Jun
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
39
|
Yin S, Zhai X, Li Y, Zeng R, Zhang D, Sun X, Zhang Z, Wang H, Wang C. Bone Metastasis Mediates Poor Prognosis in Early-Onset Gastric Cancer: Insights Into Immune Suppression, Coagulopathy, and Inflammation. Cancer Med 2025; 14:e70737. [PMID: 40040540 PMCID: PMC11880774 DOI: 10.1002/cam4.70737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/17/2025] [Accepted: 02/21/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND The increasing incidence of gastric cancer (GC) in younger populations, coupled with population aging, has highlighted distinct age-related subtypes with unique clinical characteristics and outcomes. Although younger patients tend to have more aggressive tumors, the prognostic factors for early-onset gastric cancer (EOGC) remain underexplored. This study is dedicated to providing a comprehensive and in-depth analysis of prognostic factors in EOGC, aiming to refine personalized treatment strategies under the precision medicine paradigm. METHODS This retrospective study encompassed 413 local cohort EOGC patients and 8447 Surveillance, Epidemiology, and End Results database patients diagnosed with GC. Survival outcomes were assessed using Kaplan-Meier survival curves, and differences between groups were evaluated with the log-rank test. Prognostic factors were identified through logistic regression and Cox proportional hazards models. Mediation analysis was conducted to assess the indirect effects of clinical factors on EOGC and prognosis. Biomarker comparisons between bone metastasis early-onset gastric cancer and non-bone metastasis early-onset gastric cancer groups were evaluated using the Wilcoxon test for significant differences. RESULTS The overall survival and cancer-specific survival rates in the EOGC group were significantly lower than those in the non-early-onset gastric cancer group (p < 0.05). However, EOGC itself was not an independent risk factor for poor prognosis. Mediation analysis revealed that the adverse impact of EOGC on prognosis was predominantly mediated by metastasis, with bone metastasis identified as the most significant factor. Furthermore, bone metastasis emerged as an independent predictor of poor prognosis in EOGC patients, potentially linked to elevated coagulation markers, increased inflammation-related cytokines, and an imbalance in peripheral blood immune cell ratios. CONCLUSIONS Bone metastasis significantly contributes to the poor prognosis of EOGC. EOGC patients with bone metastasis demonstrate immune suppression, inflammation activation, and coagulopathy, highlighting the need for tailored management and prognostic strategies.
Collapse
Affiliation(s)
- Shi Yin
- The Affiliated LiHuiLi Hospital of Ningbo UniversityNingboZhejiangChina
| | - Xiaohui Zhai
- Department of Medical OncologyThe Sixth Affiliated Hospital of Sun‐Yat sen UniversityGuangzhouPeople's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
- Biomedical Innovation CenterThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Yaoying Li
- State Key Laboratory of Oncology in South ChinaSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Ruixin Zeng
- Department of Medical Oncology, State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for Cancer, Sun Yat‐sen University Cancer CenterGuangzhouP. R. China
| | - Di Zhang
- Ningbo Geriatric Rehabilitation HospitalNingboZhejiangChina
| | - Xiaoqing Sun
- Department of Intensive Care Medicine (ICU), State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouP.R. China
| | - Ziying Zhang
- Department of Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunanPeople's Republic of China
| | - Huashe Wang
- Department of Gastrointestinal SurgeryThe Six Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Caiqin Wang
- Department of Lymphoma and HematologyThe Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer HospitalChangshaHunanChina
| |
Collapse
|
40
|
Zhang C, Cai H, Ye M, Wang L, Liu W, Huang Q, Peng X, Mao G, Zhang Q, Mei Y. TCF7 functions as a prognostic biomarker in bladder cancer by strengthening EMT and stemness associated with TGF-β/SMAD3 signaling. Mol Cell Biochem 2025:10.1007/s11010-025-05241-y. [PMID: 40025258 DOI: 10.1007/s11010-025-05241-y] [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: 12/02/2024] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Bladder cancer (BC) is one of the top ten most common tumors, with recurrence and metastasis being major causes of mortality among patients. A high recurrence rate is a hallmark of BC. Epithelial-mesenchymal transition (EMT) plays a role in the formation of cancer stem cells, tumor metastasis, and immune evasion. In our preliminary research, single-cell sequencing identified TCF7 as a gene associated with EMT. However, its biological function and transcriptional regulation mechanisms in BC remain unclear. This study aims to investigate TCF7's role and regulatory mechanisms in BC progression. TCF7 is a critical transcription factor promoting BC progression. High TCF7 expression in BC is significantly linked to poor patient prognosis. We uncovered a novel mechanism by which TCF7 drives EMT and stemness in BC through transcriptional regulation of TGFBR1, impacting the TGF-β/SMAD3 pathway. These findings enhance our understanding of BC progression and offer potential strategies for diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Congcong Zhang
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Haiping Cai
- Department of Neurosurgery, Guangdong Provincial People'S Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Mengnan Ye
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Le Wang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510080, Guangdong, China
| | - Weiwei Liu
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Qun Huang
- Department of Urology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Xingsi Peng
- Department of Radiation Oncology, The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, Guangzhou, 510163, Guangdong, China
| | - Guanquan Mao
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
| | - Qingling Zhang
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China.
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
| | - Yan Mei
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
41
|
Liang ZY, Yu ML, Yang H, Li HJ, Xie H, Cui CY, Zhang WJ, Luo C, Cai PQ, Lin XF, Liu KF, Xiong L, Liu LZ, Chen BY. Beyond the tumor region: Peritumoral radiomics enhances prognostic accuracy in locally advanced rectal cancer. World J Gastroenterol 2025; 31:99036. [PMID: 40062323 PMCID: PMC11886509 DOI: 10.3748/wjg.v31.i8.99036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/09/2024] [Accepted: 11/05/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND The peritumoral region possesses attributes that promote cancer growth and progression. However, the potential prognostic biomarkers in this region remain relatively underexplored in radiomics. AIM To investigate the prognostic value and importance of peritumoral radiomics in locally advanced rectal cancer (LARC). METHODS This retrospective study included 409 patients with biopsy-confirmed LARC treated with neoadjuvant chemoradiotherapy and surgically. Patients were divided into training (n = 273) and validation (n = 136) sets. Based on intratumoral and peritumoral radiomic features extracted from pretreatment axial high-resolution small-field-of-view T2-weighted images, multivariate Cox models for progression-free survival (PFS) prediction were developed with or without clinicoradiological features and evaluated with Harrell's concordance index (C-index), calibration curve, and decision curve analyses. Risk stratification, Kaplan-Meier analysis, and permutation feature importance analysis were performed. RESULTS The comprehensive integrated clinical-radiological-omics model (ModelICRO) integrating seven peritumoral, three intratumoral, and four clinicoradiological features achieved the highest C-indices (0.836 and 0.801 in the training and validation sets, respectively). This model showed robust calibration and better clinical net benefits, effectively distinguished high-risk from low-risk patients (PFS: 97.2% vs 67.6% and 95.4% vs 64.8% in the training and validation sets, respectively; both P < 0.001). Three most influential predictors in the comprehensive ModelICRO were, in order, a peritumoral, an intratumoral, and a clinicoradiological feature. Notably, the peritumoral model outperformed the intratumoral model (C-index: 0.754 vs 0.670; P = 0.015); peritumoral features significantly enhanced the performance of models based on clinicoradiological or intratumoral features or their combinations. CONCLUSION Peritumoral radiomics holds greater prognostic value than intratumoral radiomics for predicting PFS in LARC. The comprehensive model may serve as a reliable tool for better stratification and management postoperatively.
Collapse
Affiliation(s)
- Zhi-Ying Liang
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Mao-Li Yu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
- West China School of Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Hui Yang
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Hao-Jiang Li
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Hui Xie
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Chun-Yan Cui
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Wei-Jing Zhang
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Chao Luo
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Pei-Qiang Cai
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Xiao-Feng Lin
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Kun-Feng Liu
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Lang Xiong
- Department of Medical Imaging, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Li-Zhi Liu
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Bi-Yun Chen
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| |
Collapse
|
42
|
Cai R, Hong Z, Yin H, Chen H, Qin M, Huang Y. Constructing and validating a novel prognostic risk score model for rectal cancer based on four immune-related genes. Transl Cancer Res 2025; 14:1053-1069. [PMID: 40104727 PMCID: PMC11912041 DOI: 10.21037/tcr-24-1511] [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: 08/25/2024] [Accepted: 12/17/2024] [Indexed: 03/20/2025]
Abstract
Background Immunotherapy is playing an increasing role in the treatment of various cancers. However, its application in rectal cancer is very limited as only microsatellite-unstable bowel cancers with defective mismatch repair are found to benefit. The majority of rectal cancers belong to the microsatellite-stable phenotype. Therefore, the aim of this study is to explore immune-related genes within the tumor microenvironment of rectal cancer, with the objective of discovering novel biomarkers and therapeutic targets for rectal cancer, and to establish a new prognostic prediction model for rectal cancer based on these immune-related genes. Methods The data in The Cancer Genome Atlas (TCGA) database were processed using the Estimation of Stromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) algorithm to obtain differently expressed genes (DEGs). Then the DEGs were analyzed by Gene Ontology (GO), Kyoto Encyclopedia of Gene and Genomes (KEGG), Reactome function enrichment analysis, and protein-protein interaction (PPI) analysis to screen the core genes, which were utilized to compute the risk scores of individual patients. Finally, combining risk scores and clinical characteristics, a new prognostic prediction model was established by univariate and multivariate Cox analyses, and the prognostic model was validated by the Gene Expression Omnibus (GEO) database. Results The study finally identified four core genes (CYBB, CCR4, FOXP3, and CD80), and immune cell infiltration analyses of the four core genes showed that their expression levels were positively correlated with the distribution of various immune cells. The 4-gene risk score categorized rectal cancer patients into high-risk and low-risk groups, and the results showed that the low-risk group had a stronger correlation with the immune response and had a better prognosis. A prognostic model was developed by integrating risk scores and clinical characteristics and showed a strong predictive effect. Conclusions In patients with rectal cancer, CYBB, CCR4, FOXP3, and CD80 are immune-related core genes, and low expression of each gene is associated with poor clinical prognosis. The risk score obtained on their basis is independent prognostic factors for rectal cancer, suggesting that the four core genes may provide a foundation for the development of new prognostic biomarkers for rectal cancer and the study of immunotherapy.
Collapse
Affiliation(s)
- Ruyun Cai
- Department of Surgery, Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Jiaxing, China
| | - Zhonghua Hong
- Department of Surgery, Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Jiaxing, China
| | - Hezhai Yin
- Department of Surgery, Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Jiaxing, China
| | - Huilin Chen
- Department of Surgery, Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Jiaxing, China
| | - Mengting Qin
- Department of Surgery, Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Jiaxing, China
| | - Yihong Huang
- Department of Surgery, Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Jiaxing, China
| |
Collapse
|
43
|
Du X, Luo W, Li H, Gu Q, Huang P, Wang C, Li N, Liu F, Xia C. Hsa_circ_0125356 promotes gemcitabine resistance by modulating WNT canonical and non-canonical pathways via miR-582-5p/FGF9 axis in non-small cell lung cancer. Mol Cancer 2025; 24:59. [PMID: 40011917 PMCID: PMC11866803 DOI: 10.1186/s12943-025-02259-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/04/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the leading cause of cancer morbidity and mortality worldwide. The prognosis of patients has been significantly improved by chemotherapy, but acquired drug resistance remains a major obstacle to NSCLC treatment. Circular RNAs (circRNAs), which act as miRNA or protein sponges, are critically associated with the development and chemotherapy resistance of NSCLC. METHODS CircRNA sequencing was performed to analyze the differential expression of circRNAs between A549 and A549-GR cells. Chromogenic in situ hybridization (CISH) and immunohistochemistry (IHC) technologies were used to detect the expression of hsa_circ_0125356, miR-582-5p,and FGF9 in NSCLC tissues and para-carcinoma tissues. Fluorescence in situ hybridization (FISH), dual-luciferase reporter assays and RNA immunoprecipitation (RIP) were conducted to evaluate the expression and regulation of hsa_circ_0125356, miR-582-5p, and FGF9. Furthermore, the regulation of hsa_circ_0125356/miR-582-5p/FGF9 on gemcitabine sensitivity was confirmed by TUNEL, Transwell, EdU, CCK8 and immunohistochemistry. RESULTS We identified a novel hsa_circ_0125356 as a therapeutic target against gemcitabine resistance. Hsa_circ_0125356 was significantly elevated in clinical samples of patients with NSCLC. Moreover, hsa_circ_0125356 overexpression promoted gemcitabine resistance to NSCLC by upregulating FGF9 via sponging miR-582-5p in vivo and in vitro. Notably, WNT canonical (ERK/GSK3β/β-catenin) and non-canonical (Daam1/RhoA/ROCK2) signaling pathways were activated due to hsa_circ_0125356 acting as an endogenous miR-582-5p sponge to regulate the expression of FGF9, and thereby enhancing gemcitabine resistance via promoting DNA damage repair and inhibition of apoptosis. The results were further confirmed by two small molecule antagonists, WAY 316606 and XAV-939,which could inhibit the activation of WNT signaling pathway induced by hsa_circ_0125356. CONCLUSION We first demonstrated that hsa_circ_0125356 was significantly upregulated and served as a biomarker for gemcitabine resistance in NSCLC by sponging miR-582-5p/FGF9 axis to regulate the WNT canonical and non-canonical signaling pathways, which provided a new direction for identification of therapeutic targets for the treatment of gemcitabine resistance of NSCLC.
Collapse
Affiliation(s)
- Xinyue Du
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, P. R. China
| | - Weijie Luo
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, P. R. China
| | - Hongwu Li
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, P. R. China
| | - Qi Gu
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, P. R. China
| | - Ping Huang
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, P. R. China
| | - Cheng Wang
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, P. R. China
| | - Na Li
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, P. R. China
| | - Fanglan Liu
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, P. R. China.
- Key Laboratory of New Drug Transformation and Evaluation of Jiangxi Province, Nanchang, 330031, P. R. China.
| | - Chunhua Xia
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, P. R. China.
- Key Laboratory of New Drug Transformation and Evaluation of Jiangxi Province, Nanchang, 330031, P. R. China.
| |
Collapse
|
44
|
Huang H, Xu W, Feng L, Zhong ME, Ye Y, Liu Y, Ye H, Li Z, Cui Y, Liu Z, Zhao K, Yan L, Liang C. Development and evaluation of the mrTE scoring system for MRI-detected tumor deposits and extramural venous invasion in rectal cancer. Abdom Radiol (NY) 2025:10.1007/s00261-025-04840-z. [PMID: 39954064 DOI: 10.1007/s00261-025-04840-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/03/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
PURPOSE Tumor deposits (TDs) and extramural venous invasion (EMVI) in locally advanced rectal cancer (LARC) are MRI-detectable markers that reflect the invasive and metastatic potential of tumors. However, both mrTDs and mrEMVI are closely associated with peritumoral vascular signals, and they show a high degree of statistical correlation. We developed a novel scoring system that integrates mrTDs and mrEMVI into a single parameter, simplifying the assessment process and capturing the prognostic value of both factors simultaneously. METHODS We retrospectively included LARC patients who received neoadjuvant chemoradiotherapy at five centers and proposed a novel MRI-based scoring system, mrTE (derived from mrTDs and mrEMVI), to integrate the prognostic significance of mrEMVI and mrTDs in rectal cancer. The prognostic value of different mrTE scores was evaluated using Kaplan-Meier curves and the Cox model. The predictive accuracy of the new scoring system was evaluated using the integrated area under the ROC curve (iAUC). RESULTS A total of 1188 patients with LARC were included in the evaluation cohort to assess the reliability of the novel imaging scoring system. Based on the mrTE scores ranging from 0 to 2, the patients were categorized into three groups. The 3-year disease-free survival rates for the groups were 88.1%, 78.1%, and 51.9% (score 1 vs 0: HR 2.00, 95% CI 1.36-2.93, p < 0.001; score 2 vs 0: HR 4.75, 95% CI 3.61-6.26, p < 0.001). The mrTE scoring system demonstrated superior performance in predicting DFS compared to other clinical and imaging markers, with a higher predictive accuracy (iAUC = 0.707). CONCLUSIONS The mrTE scoring system simplifies the clinical assessment of relevant MR markers and has proven to be an effective tool for predicting the prognosis of LARC patients.
Collapse
Affiliation(s)
- Haitao Huang
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Weixiong Xu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lili Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Min-Er Zhong
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, PR China
| | - Yunrui Ye
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yulin Liu
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huifen Ye
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesFudan University Shanghai Cancer Center, Shanghai, China
| | - Zhenhui Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Yanfen Cui
- Department of Radiology, Shanxi Cancer Hospital, Shanxi Medical University, Taiyuan, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Ke Zhao
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China.
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Lifen Yan
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Changhong Liang
- School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China.
| |
Collapse
|
45
|
Morshidi NAAB, Uddin MS, Lee J, Han SI, Kim JH. Anticancer activity of Trigonella Foenumgraecum (fenugreek) seed extract by inducing apoptosis in pancreatic cancer cell. Am J Transl Res 2025; 17:832-843. [PMID: 40092083 PMCID: PMC11909546 DOI: 10.62347/pglt6191] [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: 08/13/2024] [Accepted: 01/18/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND/AIM Pancreatic cancer exhibits resistance to currently available drugs in the pharmaceutical industry. The development of new drugs is crucial, and research on plant substances with biological activities against cancer is actively underway. This study explored the potential use of fenugreek seed extract (FSE) in pancreatic cancer treatment as the anticancer activity of FSE is still poorly understood. MATERIALS AND METHODS The anticancer activity of FSE on pancreatic cancer cells was evaluated using cell viability and apoptosis assays. The migration rate of cancer cells was quantified using wound healing and Transwell migration assays. Western blotting was utilized to assess relevant signaling pathways, and LC-MS/MS was employed to detect the active compounds in FSE. RESULTS FSE inhibited the proliferation of pancreatic cancer cell lines (Panc-1, Miapaca-2, SNU-213, and Aspc-1) in a time and dose-dependent manner without greatly affecting normal cells (293T). The inhibition of cancer cell proliferation was attributed to the activation of cleaved caspase-3 and Bax, a pro-apoptotic marker. The anticancer effects and inhibition of cell migration were mediated by the MAPK, Akt, MMP-9, and vimentin signaling pathways through inactivation of the phosphorylated proteins related to cell growth, differentiation, and migration. LC-MS/MS analysis detected various active compounds capable of inducing apoptosis in pancreatic cancer cells. CONCLUSION We demonstrated that FSE has anticancer properties by inducing apoptosis and preventing metastasis in pancreatic cancer cells without affecting normal cells.
Collapse
Affiliation(s)
| | - Md Salah Uddin
- Botanical Research Centre Botanika, Tejgaon, Dhaka 1208, Bangladesh
| | - Jungwhoi Lee
- Subtropical/Tropical Organism Gene Bank, Jeju National University Jeju 63608, Republic of Korea
| | - Song-I Han
- Subtropical/Tropical Organism Gene Bank, Jeju National University Jeju 63608, Republic of Korea
| | - Jae-Hoon Kim
- Department of Biotechnology, College of Applied Life Science, SARI, Jeju National University Jeju 63608, Republic of Korea
- Subtropical/Tropical Organism Gene Bank, Jeju National University Jeju 63608, Republic of Korea
| |
Collapse
|
46
|
Yang H, Zhu J, Wang X. Comprehensive Analysis Identifies Hsa_circ_0058191 as a Potential Drug Resistance Target in Multiple Myeloma. Onco Targets Ther 2025; 18:225-231. [PMID: 39963489 PMCID: PMC11831480 DOI: 10.2147/ott.s505074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/31/2025] [Indexed: 02/20/2025] Open
Abstract
Background Multiple Myeloma (MM) is the second most common hematologic malignancy, which exhibits strong resistance to bortezomib, the first-line treatment. Circular RNAs (circRNAs) are increasingly considered as important drivers of drug resistance across various cancers, but their roles in multiple myeloma are not well understood. Aim To investigate and identify potential circRNA targets and their roles in the mechanisms of bortezomib resistance. Methods Bortezomib-resistant MM patient-specific circRNAs were screened using Arraystar circRNA microarrays. The MM circRNA dataset from the GEO database was analyzed with GEO2R to identify candidate circRNAs associated with MM progression and drug resistance. CircRNA-forming and loop-forming sites, along with their structures, were identified via Sanger sequencing. The identified circRNA was validated by qRT-PCR in MM patients with and without bortezomib resistance. Bioinformatic analysis through CircInteractome was conducted to predict potential miRNA and RBP binding for the core circRNAs. Metascape was employed to perform RBP pathway analysis to identify specific biological processes in circRNAs. Results The hsa_circ_0058191 was found to be overexpressed in bortezomib-resistant MM patient samples, suggesting its pivotal role in drug resistance mechanisms. The interaction of hsa_circ_0058191 with miR-660 and AGO2 as determined through bioinformatic predictions, indicated that it regulates RNA modification and mRNA regulation pathways. These molecular interactions expand our understanding of the mechanisms of drug resistance in multiple myeloma. Conclusion This study identified the role of hsa_circ_0058191 in the development of drug resistance in MM, which provides a theoretical foundation for designing potential therapeutic strategies to prevent drug resistance.
Collapse
Affiliation(s)
- Huiye Yang
- Department of Hematology, The Affiliated Hospital of Guilin Medical University, Guilin, People’s Republic of China
| | - Jie Zhu
- Department of Hematology, The Affiliated Hospital of Guilin Medical University, Guilin, People’s Republic of China
| | - Xiaotao Wang
- Department of Hematology, The Affiliated Hospital of Guilin Medical University, Guilin, People’s Republic of China
| |
Collapse
|
47
|
Luo D, Zhou J, Ruan S, Zhang B, Zhu H, Que Y, Ying S, Li X, Hu Y, Song Z. Overcoming immunotherapy resistance in gastric cancer: insights into mechanisms and emerging strategies. Cell Death Dis 2025; 16:75. [PMID: 39915459 PMCID: PMC11803115 DOI: 10.1038/s41419-025-07385-7] [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: 11/07/2024] [Revised: 01/07/2025] [Accepted: 01/22/2025] [Indexed: 02/09/2025]
Abstract
Gastric cancer (GC) remains a leading cause of cancer-related mortality worldwide, with limited treatment options in advanced stages. Immunotherapy, particularly immune checkpoint inhibitors (ICIs) targeting PD1/PD-L1, has emerged as a promising therapeutic approach. However, a significant proportion of patients exhibit primary or acquired resistance, limiting the overall efficacy of immunotherapy. This review provides a comprehensive analysis of the mechanisms underlying immunotherapy resistance in GC, including the role of the tumor immune microenvironment, dynamic PD-L1 expression, compensatory activation of other immune checkpoints, and tumor genomic instability. Furthermore, the review explores GC-specific factors such as molecular subtypes, unique immune evasion mechanisms, and the impact of Helicobacter pylori infection. We also discuss emerging strategies to overcome resistance, including combination therapies, novel immunotherapeutic approaches, and personalized treatment strategies based on tumor genomics and the immune microenvironment. By highlighting these key areas, this review aims to inform future research directions and clinical practice, ultimately improving outcomes for GC patients undergoing immunotherapy.
Collapse
Affiliation(s)
- Dingtian Luo
- Gastroenterology Department, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jing Zhou
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Shuiliang Ruan
- Gastroenterology Department, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Binzhong Zhang
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Huali Zhu
- Gastroenterology Department, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Yangming Que
- Gastroenterology Department, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Shijie Ying
- Gastroenterology Department, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Xiaowen Li
- Pathology Department, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Yuanmin Hu
- Intensive Care Unit, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| | - Zhengwei Song
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| |
Collapse
|
48
|
Bora Yildiz C, Du J, Mohan KN, Zimmer-Bensch G, Abdolahi S. The role of lncRNAs in the interplay of signaling pathways and epigenetic mechanisms in glioma. Epigenomics 2025; 17:125-140. [PMID: 39829063 PMCID: PMC11792803 DOI: 10.1080/17501911.2024.2442297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Gliomas, highly aggressive tumors of the central nervous system, present overwhelming challenges due to their heterogeneity and therapeutic resistance. Glioblastoma multiforme (GBM), the most malignant form, underscores this clinical urgency due to dismal prognosis despite aggressive treatment regimens. Recent advances in cancer research revealed signaling pathways and epigenetic mechanisms that intricately govern glioma progression, offering multifaceted targets for therapeutic intervention. This review explores the dynamic interplay between signaling events and epigenetic regulation in the context of glioma, with a particular focus on the crucial roles played by non-coding RNAs (ncRNAs). Through direct and indirect epigenetic targeting, ncRNAs emerge as key regulators shaping the molecular landscape of glioblastoma across its various stages. By dissecting these intricate regulatory networks, novel and patient-tailored therapeutic strategies could be devised to improve patient outcomes with this devastating disease.
Collapse
Affiliation(s)
- Can Bora Yildiz
- Division of Neuroepigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, Aachen, Germany
- Research Training Group 2416 Multi Senses – Multi Scales, RWTH Aachen University, Aachen, Germany
| | - Jian Du
- Division of Neuroepigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, Aachen, Germany
| | - K. Naga Mohan
- Molecular Biology and Genetics Laboratory, Department of Biological Sciences, Hyderabad, India
| | - Geraldine Zimmer-Bensch
- Division of Neuroepigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, Aachen, Germany
- Research Training Group 2416 Multi Senses – Multi Scales, RWTH Aachen University, Aachen, Germany
| | - Sara Abdolahi
- Division of Neuroepigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, Aachen, Germany
| |
Collapse
|
49
|
Zhou J, Li L, Liu Y, Jia W, Liu Q, Gao X, Wu A, Wu B, Shen Z, Wang Z, Han J, Niu B, Gong Y, Guan Y, Zhou J, Xue H, Zhou W, Hu K, Lu J, Xu L, Xia X, Yi X, Yang L, Lin G. Circulating tumour DNA in predicting and monitoring survival of patients with locally advanced rectal cancer undergoing multimodal treatment: long-term results from a prospective multicenter study. EBioMedicine 2025; 112:105548. [PMID: 39818166 PMCID: PMC11786667 DOI: 10.1016/j.ebiom.2024.105548] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/22/2024] [Accepted: 12/22/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Neoadjuvant chemoradiotherapy (nCRT) is the standard for locally advanced rectal cancer (LARC). However, distant metastasis remains the primary cause of treatment failure. Early identification of high-risk individuals for personalized treatment may offer a solution. Circulating tumour DNA (ctDNA) could assist in this process. METHODS From September 2017 to June 2019, the study prospectively recruited 113 patients with LARC (cT3-4N0M0 or cTanyN + M0) who underwent nCRT followed by radical surgery across 8 tertiary centers. ctDNA was analysed using large-panel targeted sequencing at baseline, during nCRT, pre-surgery, post-surgery, post-adjuvant chemotherapy (ACT), and during annual follow-ups for 3 years. FINDINGS We analysed 103 tissue and 669 plasma samples from 103 patients. With a median 53-month follow-up, significantly worse progression-free survival (PFS) and overall survival (OS) were observed if median variant allele frequency (mVAF) of baseline ctDNA per patient was ≥0.5% (PFS, HR 4.39, p < 0.001; OS, HR 5.61, p = 0.004) or ctDNA was still detectable two weeks into nCRT (PFS, HR 7.63, p < 0.001; OS, HR 5.08, p < 0.001). Furthermore, when compared to the low-risk (C1) group (characterized by "ctDNA undetected during nCRT with baseline mVAF <0.5%" or "ctDNA undetected during nCRT with TMB (tumour mutational burden) ≥20/Mb"), the high-risk (C2) group (characterized by "ctDNA detected during nCRT" or "baseline mVAF ≥0.5% with TMB <20/Mb") showed significantly worse long-term outcomes (3 y-PFS, 55.9% vs. 94.2%; 3 y-OS, 79.4% vs. 100%). The ctDNA clearance during nCRT, baseline mVAF, and TMB may be effective prognostic indicators. INTERPRETATION Our findings reaffirm the clinical monitoring value of ctDNA and demonstrate the strong prognostic value of baseline ctDNA and its early clearance status in patients with LARC undergoing nCRT. This highlights the potential of dynamic ctDNA monitoring as actionable stratified indicators to guide personalized neoadjuvant treatment strategies. FUNDING This work was supported by the Major Grants Program of Beijing Science and Technology Commission (No. D171100002617003) and the National High Level Hospital Clinical Research Funding (2022-PUMCH-C-005).
Collapse
Affiliation(s)
- Jiaolin Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Lifeng Li
- Geneplus-Beijing, Beijing 102206, China
| | - Yuxin Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Wenzhuo Jia
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Qian Liu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xuan Gao
- Geneplus-Beijing, Beijing 102206, China
| | - Aiwen Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Unit III, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Bin Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Zhanlong Shen
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100871, China
| | - Zhenjun Wang
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jiagang Han
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Beizhan Niu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | | | | | - Jianfeng Zhou
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Huadan Xue
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Weixun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Ke Hu
- Department of Radiotherapy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Junyang Lu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Lai Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | | | - Xin Yi
- Geneplus-Beijing, Beijing 102206, China
| | - Ling Yang
- Geneplus-Beijing, Beijing 102206, China.
| | - Guole Lin
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
50
|
Chaudhary JK, Danga AK, Kumari A, Bhardwaj A, Rath PC. Role of chemokines in aging and age-related diseases. Mech Ageing Dev 2025; 223:112009. [PMID: 39631472 DOI: 10.1016/j.mad.2024.112009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Chemokines (chemotactic cytokines) play essential roles in developmental process, immune cell trafficking, inflammation, immunity, angiogenesis, cellular homeostasis, aging, neurodegeneration, and tumorigenesis. Chemokines also modulate response to immunotherapy, and consequently influence the therapeutic outcome. The mechanisms underlying these processes are accomplished by interaction of chemokines with their cognate cell surface G protein-coupled receptors (GPCRs) and subsequent cellular signaling pathways. Chemokines play crucial role in influencing aging process and age-related diseases across various tissues and organs, primarily through inflammatory responses (inflammaging), recruitment of macrophages, and orchestrated trafficking of other immune cells. Chemokines are categorized in four distinct groups based on the position and number of the N-terminal cysteine residues; namely, the CC, CXC, CX3C, and (X)C. They mediate inflammatory responses, and thereby considerably impact aging process across multiple organ-systems. Therefore, understanding the underlying mechanisms mediated by chemokines may be of crucial importance in delaying and/or modulating the aging process and preventing age-related diseases. In this review, we highlight recent progress accomplished towards understanding the role of chemokines and their cellular signaling pathways involved in aging and age-relaed diseases of various organs. Moreover, we explore potential therapeutic strategies involving anti-chemokines and chemokine receptor antagonists aimed at reducing aging and mitigating age-related diseases. One of the modern methods in this direction involves use of chemokine receptor antagonists and anti-chemokines, which suppress the pro-inflammatory response, thereby helping in resolution of inflammation. Considering the wide-spectrum of functional involvements of chemokines in aging and associated diseases, several clinical trials are being conducted to develop therapeutic approaches using anti-chemokine and chemokine receptor antagonists to improve life span and promote healthy aging.
Collapse
Affiliation(s)
- Jitendra Kumar Chaudhary
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India; Department of Zoology, Shivaji College, University of Delhi, New Delhi 110027, India.
| | - Ajay Kumar Danga
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Anita Kumari
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Akshay Bhardwaj
- Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad Road, Faridabad, Haryana 121001, India.
| | - Pramod C Rath
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|