1
|
Huang FJ, Fang YY, Wen JY, Li JJ, Lin Q, Su QY, Chen YY, Wang L, Zeng JJ, Chi BT, He RQ, Qin DY, Yang LH, Chen G. From PD-1/PD-L1 to tertiary lymphoid structures: Paving the way for precision immunotherapy in cholangiocarcinoma treatment. Hum Vaccin Immunother 2025; 21:2444697. [PMID: 39757910 DOI: 10.1080/21645515.2024.2444697] [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: 09/17/2024] [Revised: 12/07/2024] [Accepted: 12/16/2024] [Indexed: 01/07/2025] Open
Abstract
Cholangiocarcinoma (CCA) is a highly malignant hepatobiliary tumor characterized by limited treatment options and poor prognosis. The recent rise of immunotherapy has significantly influenced research in this field. This study presents a bibliometric analysis of 416 articles retrieved from the WOSCC, Wan fang Data, CNKI and VIP databases, spanning contributions from 32 countries, 589 institutions and 3,200 authors. The analysis identified "PD-L1," "PD-1" and "pembrolizumab" as central research foci, while "immune checkpoint inhibitors," "tumor immune microenvironment," "tertiary lymphoid structures" and "durvalumab" emerged as key areas of interest. These findings emphasize the pivotal role of immunotherapy in improving survival outcomes for CCA, and they highlight the significance of tertiary lymphoid structures within the tumor microenvironment as a promising target for future research. This study offers a strategic overview of the evolving landscape of CCA immunotherapy, providing valuable insights to guide future scientific endeavors in this domain.
Collapse
Affiliation(s)
- Fang-Ju Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Ye-Ying Fang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Jia-Ying Wen
- Department of Radiotherapy, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Jian-Jun Li
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Qian Lin
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Qin-Yan Su
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Yi-Yang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Lei Wang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Jian-Jia Zeng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Bang-Teng Chi
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Di-Yuan Qin
- Department of Computer Science and Technology, School of Computer and Electronic Information, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Li-Hua Yang
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| |
Collapse
|
2
|
Duan Y, Liu Z, Wang Q, Zhang J, Liu J, Zhang Z, Li C. Targeting MYC: Multidimensional regulation and therapeutic strategies in oncology. Genes Dis 2025; 12:101435. [PMID: 40290126 PMCID: PMC12022651 DOI: 10.1016/j.gendis.2024.101435] [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: 04/16/2024] [Revised: 08/05/2024] [Accepted: 08/25/2024] [Indexed: 04/30/2025] Open
Abstract
MYC is dysregulated in approximately 70% of human cancers, strongly suggesting its essential function in cancer. MYC regulates many biological processes, such as cell cycle, metabolism, cellular senescence, apoptosis, angiogenesis, and immune escape. MYC plays a central role in carcinogenesis and is a key regulator of tumor development and drug resistance. Therefore, MYC is one of the most alluring therapeutic targets for developing cancer drugs. Although the search for direct inhibitors of MYC is challenging, MYC cannot simply be assumed to be undruggable. Targeting the MYC-MAX complex has been an effective method for directly targeting MYC. Alternatively, indirect targeting of MYC represents a more pragmatic therapeutic approach, mainly including inhibition of the transcriptional or translational processes of MYC, destabilization of the MYC protein, and blocking genes that are synthetically lethal with MYC overexpression. In this review, we delineate the multifaceted roles of MYC in cancer progression, highlighting a spectrum of therapeutic strategies and inhibitors for cancer therapy that target MYC, either directly or indirectly.
Collapse
Affiliation(s)
- Yingying Duan
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Zhaoshuo Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Qilin Wang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Junyou Zhang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Jiaxin Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Ziyi Zhang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Chunyan Li
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing 100191, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, China
| |
Collapse
|
3
|
Chen W, Zhang G, Lei Q, Lu H. Prognostic value of advanced lung cancer inflammation index in heart failure patients: A comprehensive analysis. ESC Heart Fail 2025; 12:2298-2309. [PMID: 39624970 PMCID: PMC12055415 DOI: 10.1002/ehf2.15178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/18/2024] [Accepted: 11/08/2024] [Indexed: 05/08/2025] Open
Abstract
AIMS The prognosis of heart failure (HF) is closely linked to inflammation and nutritional status. The advanced lung cancer inflammation index (ALI) is a composite indicator consisting of several parameters used to assess inflammation and nutritional status. Our study aimed to investigate the prognostic value of ALI in HF patients. METHODS The data from Study 1, which included 1359 HF patients, were extracted from the National Health and Nutrition Examination Survey (NHANES) database spanning the years 1999 to 2018. Study 2 analysed data from patients with HF who underwent cardiac magnetic resonance imaging examinations from 2020 to 2023. Kaplan-Meier curve analysis, Cox proportional hazard model, time-dependent receiver operating characteristic (ROC) curve and restricted cubic spline (RCS) were used to evaluate the relationship between ALI and long-term prognosis of patients with HF in Study 1. Logistic regression analysis was used to evaluate the correlation between ALI and left ventricular reverse remodelling, and RCS was used to determine any dose-response relationship. Spearman correlation was used to evaluate the relationship between ALI and indicators of cardiac structural changes. RESULTS Study 1 found that the average age of the patients was 68 years [inter-quartile range (IQR) 58-76], the proportion of males was 54.3%, and there were 699 all-cause mortality and 293 cardiovascular mortality cases. After adjusted by multivariable Cox regression analysis, elevated ALI levels were significantly associated with increased risks of all-cause [hazard ratio (HR) = 0.58, 95% confidence interval (CI) = 0.42-0.79, P < 0.001] and cardiovascular mortality (HR = 0.61, 95% CI = 0.38-0.97, P = 0.036) in patients with HF. A linear negative correlation was observed between ALI and both all-cause (P = 0.0011 and P < 0.001, P for nonlinear = 0.3993) and cardiovascular mortality (P = 0.0011, P for nonlinear = 0.5198). Time-dependent ROC curves showed the predictive value of ALI for all-cause mortality [area under the curve (AUC) = 0.678 in 3 years, AUC = 0.674 in 5 years and AUC = 0.683 in 10 years] and cardiovascular mortality (AUC = 0.694 in 3 years, AUC = 0.685 in 5 years and AUC = 0.697 in 10 years). Study 2 included 79 patients; the average age of the patients was 44 years (IQR 35-55); and the proportion of males was 74.7%. Adjusted multivariable logistic regression analysis indicated that high ALI levels were associated with left ventricular reverse remodelling (LVRR) in patients with HF following discharge from the hospital [odds ratio (OR) = 3.16, 95% CI = 1.06-10.8, P = 0.049]. Spearman analysis revealed a correlation between ALI and extracellular volume (ECV) (r = -0.25, P = 0.023). CONCLUSION ALI is associated with all-cause and cardiovascular mortality risk and structural changes in the heart in patients with HF.
Collapse
Affiliation(s)
- Wentao Chen
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Guihua Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Qiao Lei
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Huixia Lu
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| |
Collapse
|
4
|
Chen L, Li Q. Nanomaterials in the diagnosis and treatment of gastrointestinal tumors: New clinical choices and treatment strategies. Mater Today Bio 2025; 32:101782. [PMID: 40331152 PMCID: PMC12051065 DOI: 10.1016/j.mtbio.2025.101782] [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: 02/26/2025] [Revised: 04/14/2025] [Accepted: 04/18/2025] [Indexed: 05/08/2025] Open
Abstract
Nanomaterials have emerged as a promising modality in the diagnosis and treatment of gastrointestinal (GI) tumors, offering significant advancements over conventional methods. In diagnostic applications, nanomaterials facilitate enhanced imaging techniques, including magnetic resonance imaging (MRI), computed tomography (CT), and fluorescence imaging, which provide improved resolution and more accurate detection of early-stage cancers. Nanoparticles (NPs), such as liposomes, dendrimers, and quantum dots, are increasingly employed for the targeted imaging of specific biomarkers associated with GI malignancies, thereby enhancing diagnostic sensitivity and specificity. Liposomes are primarily used for drug delivery due to their ability to encapsulate hydrophobic drugs, dendrimers are useful for both drug delivery and gene therapy due to their highly branched structure, and quantum dots are primarily used in imaging and diagnostics because of their fluorescent properties. We also discuss their respective advantages and limitations. In therapeutic contexts, nanomaterials play a pivotal role in the development of targeted drug delivery systems. These systems address the limitations of traditional chemotherapy by improving drug bioavailability, reducing systemic toxicity, and promoting selective accumulation at tumor sites via both passive and active targeting mechanisms. Nanomedicines, including NPs and nanocarriers, enable the precise delivery of chemotherapeutic agents, nucleic acid -based therapies, and immunomodulators directly to cancer cells, thereby optimizing therapeutic efficacy. Furthermore, nanotechnology offers the potential to modulate the tumor microenvironment (TME), a critical factor in overcoming challenges related to tumor resistance and metastasis. Despite these promising advancements, several challenges persist, including concerns regarding long-term toxicity, stability, and regulatory approval. Nonetheless, the integration of nanomaterials into clinical practice holds substantial potential for revolutionizing the management of GI cancers, paving the way for more precise, personalized, and effective therapeutic strategies.
Collapse
Affiliation(s)
- Liping Chen
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, PR China
| | - Qingqing Li
- Department of Endoscopy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, PR China
| |
Collapse
|
5
|
Zhao H, Mao Y, Wang H, Zhou A, Yang Z, Han Y, Li G, Bi X, Hao C, Wang X, Zhou J, Dai C, Wen F, Zhang J, Liu R, Li T, Zhao L, Niu Z, Wen T, Li Q, Zhang H, Chen X, Chen M, Zhao M, Chen Y, Yu J, Shen J, Li X, Liu L, Huang Z, Zhang W, Shen F, Zhou W, Yuan Z, Zhai J, Ge N, Chen Y, Sun H, Cai J. A Survey of Clinical Practices for Hepatocellular Carcinoma Among Experts at Tertiary Hospitals in China From 2020 to 2021. CANCER INNOVATION 2025; 4:e70006. [PMID: 40196745 PMCID: PMC11975463 DOI: 10.1002/cai2.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/08/2024] [Accepted: 08/03/2024] [Indexed: 04/09/2025]
Abstract
Background Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related death in China. The rapid progress in systemic therapies has led to the approval of many therapeutic methods that have quickly changed clinical guidelines and practices. Because of the high heterogeneity of HCC, there are still some gaps between the guidelines and real-world clinical practice. The present study surveyed experts in China to investigate the current treatment concepts and clinical practice regarding HCC. Methods A questionnaire survey on the treatment concepts and clinical practice of HCC was administered to 310 experts with senior professional titles in 2020 and 312 experts in 2021. The results were analyzed and compared. Results For treating patients with resectable HCC, 28% of hepatobiliary surgeons indicated neoadjuvant therapy, and 7% chose systemic therapy ± locoregional therapy as 1 L therapy in 2021 compared with 20% and 1% in 2020. More experts chose adjuvant treatment within 1 month in 2021 compared with 2020, and 6 months and 12 months were the leading choices for the duration of adjuvant treatment. In 2021, 79% of surgeons and 19% of interventionalists were willing to conduct downstaging/conversion therapy for patients with potentially resectable HCC, and 78% chose tyrosine kinase inhibitors (TKI) + immunotherapy (IO) + locoregional therapy for cases in which R0 resection could not be achieved. For completely unresectable HCC, more experts preferred TKI + IO-based therapy as 1 L therapy in 2021 compared with 2020 (78% vs. 55%). The proportion of experts who indicated TKI + IO-based therapy as 2 L therapy increased from 32% in 2020 to 40% in 2021. Conclusion The survey results indicated that in 2021, compared with 2020, more experts opted to administer IO + TKI for the treatment of liver cancer, and more experts and patients were willing to participate in clinical research.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) HospitalPUMC and Chinese Academy of Medical Sciences (CAMS)BeijingChina
| | - Hongguang Wang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Aiping Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhengqiang Yang
- Department of Interventional Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yue Han
- Department of Interventional Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Gong Li
- Department of Radiation Oncology, Beijing Tsinghua Changgung Hospital (BTCH)School of Clinical Medicine, Tsinghua UniversityBeijingChina
| | - Xinyu Bi
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Chunyi Hao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Sarcoma CenterPeking University Cancer Hospital & InstituteBeijingChina
| | - Xiaodong Wang
- Departments of Interventional OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Jun Zhou
- Department of Medical OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Chaoliu Dai
- Department of General SurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Feng Wen
- Department of RadiologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jingdong Zhang
- Medical Oncology Department of Gastrointestinal CancerLiaoning Cancer Hospital & Institute, Cancer Hospital of China Medical UniversityShenyangLiaoningChina
| | - Ruibao Liu
- Interventional Radiological DepartmentHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Tao Li
- Department of General Surgery, Qilu HospitalThe Second Hospital of Shandong UniversityJinanShandongChina
| | - Lei Zhao
- Department of Hepatobiliary SurgeryShandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical ScienceJinanShandongChina
| | - Zuoxing Niu
- Department of Gastroenterology, Ward 2, Shandong Cancer Hospital and InstituteShandong First Medical UniversityJinanShandongChina
| | - Tianfu Wen
- Department of Liver Surgery, West China HospitalSichuan UniversityChengduSichuanChina
| | - Qiu Li
- Cancer Center, West China HospitalSichuan UniversityChengduSichuanChina
| | - Hongmei Zhang
- Department of Clinical Oncology, Xijing HospitalThe Air Force Military Medical UniversityXi'anShaanxiChina
| | - Xiaoming Chen
- Department of Interventional RadiologyGuangdong Provincial People's HospitalGuangzhouGuangdongChina
| | - Minshan Chen
- Department of Liver SurgerySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdongChina
| | - Ming Zhao
- Department of Minimally Invasive Interventional Therapy, Liver Cancer Study and Service GroupSun Yat‐sen University Cancer CenterGuangzhouGuangdongChina
| | - Yajin Chen
- Department of Hepatobiliary Surgery, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Jun Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
| | - Jie Shen
- Department of OncologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
| | - Xiangchen Li
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Zhiyong Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Wei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Feng Shen
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery HospitalSecond Military Medical University (Naval Medical University)ShanghaiChina
| | - Weiping Zhou
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery HospitalSecond Military Medical University (Naval Medical University)ShanghaiChina
| | - Zhengang Yuan
- Department of Oncology, Eastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| | - Jian Zhai
- Department II of Interventional RadiologyEastern Hepatobiliary Surgery HospitalShanghaiChina
| | - Ningling Ge
- Department of Hepatic Oncology, Zhongshan Hospital, Liver Cancer Institute and Key Laboratory of Carcinogenesis and Cancer InvasionFudan UniversityShanghaiChina
| | - Yongjun Chen
- Department of General Surgery, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Huichuan Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan HospitalFudan UniversityShanghaiChina
| | - Jianqiang Cai
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
6
|
Nair RS, Patel MN, Kannan T, Gour S, Hariharan MM, Prasanna V, Thirumalai A, Chockalingam R, Vasantharekha R, ThyagaRajan S, Priyanka HP. Effects of 17β-estradiol and estrogen receptor subtype-specific agonists on Jurkat E6.1 T-cell leukemia cells. Toxicol In Vitro 2025; 106:106057. [PMID: 40112934 DOI: 10.1016/j.tiv.2025.106057] [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: 12/16/2024] [Revised: 02/23/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Estrogen signaling plays a crucial role in immune regulation and cancer metabolism, yet its impact on T-cell leukemia remains unclear. In hematological malignancies, estrogen receptor (ER) activation may influence metabolic shifts that affect cell survival and proliferation. This study investigates the in vitro effects of 17β-estradiol and estrogen receptor subtype-specific agonists on Jurkat E6.1 T-cell leukemia cells. PURPOSE To assess how estrogen signaling influences metabolic reprogramming, inflammatory response, and survival pathways in Jurkat E6.1 cells through receptor-dependent and independent mechanisms. METHODS Jurkat E6.1 cells incubated with different concentrations of 17β-estradiol (10-12 M, 10-10 M, 10-8 M) or ER-α agonist 4,4',4″-(4-Propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (10-10 M, 10-8 M, 10-6 M) or ER-β agonist diarylproprionitrile (10-10 M, 10-8 M, 10-6 M) with and without non-specific antagonist ICI 182,780 (10-6 M). The metabolic enzyme activities of hexokinase, pyruvate kinase, and citrate synthase were measured in cell pellets, while supernatants were analyzed for IL-6 and nitric oxide (NO) production. Additionally, PI3K/Akt pathway activation was assessed by measuring p-Akt/Total Akt expression. RESULTS A shift from glycolysis to oxidative phosphorylation was observed on treatment with 17β-estradiol with significant decline in hexokinase activity and a concomitant increase in activities of pyruvate kinase and citrate synthase. CONCLUSION 17β-estradiol mediates its effects on Jurkat E6.1 cells in vitro through receptor-subtype dependent and independent mechanisms involving metabolic enzymes (hexokinase, pyruvate kinase, citrate synthase), cytokines (IL-6), nitric oxide, and signaling molecules (p-Akt).
Collapse
Affiliation(s)
- Rahul S Nair
- Inspire Laboratory, Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Chennai, Tamil Nadu, India; Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Mantavya N Patel
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Thangamani Kannan
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Shaili Gour
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Murali M Hariharan
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Vijayarengamani Prasanna
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Anupriya Thirumalai
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Ramanathan Chockalingam
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Ramasamy Vasantharekha
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Srinivasan ThyagaRajan
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Hannah P Priyanka
- Inspire Laboratory, Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Chennai, Tamil Nadu, India; Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India.
| |
Collapse
|
7
|
Emmen AMLH, Ali M, Groot Koerkamp B, Boggi U, Molenaar IQ, Busch OR, Hackert T, Moraldi L, Mieog JS, Lips DJ, Saint-Marc O, Luyer MDP, van Dieren S, Kazemier G, Nickel F, Festen S, van Santvoort HC, Kauffmann EF, de Wilde RF, Abu Hilal M, Besselink MG. Predicting postoperative pancreatic fistula after robotic pancreatoduodenectomy using International Study Group on Pancreatic Surgery and fistula risk scores: European multicentre retrospective cohort study. BJS Open 2025; 9:zraf036. [PMID: 40331890 DOI: 10.1093/bjsopen/zraf036] [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: 07/08/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Postoperative pancreatic fistula represents the leading cause of morbidity and mortality after robotic pancreatoduodenectomy. Various scores have been proposed to stratify patients based on their postoperative pancreatic fistula risk, including three fistula risk scores, and two International Study Group for Pancreatic Surgery scores. This study compares the performance of these scores in patients undergoing robotic pancreatoduodenectomy. METHODS This is a multicentre European retrospective study in consecutive patients receiving robotic pancreatoduodenectomy for all indications (April 2014 to December 2021). The performance of the International Study Group for Pancreatic Surgery 4-tier (A-D) risk score, and its 3-tier (A-C) modification (International Study Group for Pancreatic Surgery 3-tier), fistula risk scores, alternative-fistula risk scores and the updated alternative-fistula risk scores in postoperative pancreatic fistula grade B/C prediction were compared based on their discrimination (area under the curve), calibration and clinical utility, evaluated through decision curve analyses. RESULTS Overall, 919 patients undergoing robotic pancreatoduodenectomy were included. The rate of grade B/C postoperative pancreatic fistula was 22.2% (n = 204). The area under the curve for the five scores differed only slightly: International Study Group for Pancreatic Surgery 0.63 (95% confidence interval (c.i.) 0.58 to 0.67), International Study Group for Pancreatic Surgery 3-tier 0.63 (95% c.i. 0.58 to 0.67), fistula risk scores 0.65 (95% c.i. 0.61 to 0.69), alternative-fistula risk scores 0.64 (95% c.i. 0.60 to 0.68) and updated alternative-fistula risk scores 0.65 (95% c.i. 0.60 to 0.69). The International Study Group for Pancreatic Surgery, International Study Group for Pancreatic Surgery 3-tier, fistula risk scores and alternative-fistula risk scores underestimated the risk of postoperative pancreatic fistula. In contrast, the updated alternative-fistula risk score was well-calibrated at low predicted risks, but overestimated postoperative pancreatic fistula risk for high-risk patients. In decision curve analyses, the updated alternative-fistula risk score showed a higher clinical utility compared with the four other risk scores. CONCLUSION The clinical utility of the updated alternative-fistula risk score for robotic pancreatoduodenectomy slightly outperformed the four other fistula risk scores, and might be used for patient counselling and patient stratification in clinical practice and research.
Collapse
Affiliation(s)
- Anouk M L H Emmen
- Department of Surgery, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Mahsoem Ali
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Ugo Boggi
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| | - I Quintus Molenaar
- Department of Surgery, Regional Academic Cancer Centre Utrecht, St. Antonius Hospital and University Medical Centre, Utrecht, The Netherlands
| | - Olivier R Busch
- Department of Surgery, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Thilo Hackert
- Dept. of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Luca Moraldi
- Department of Oncology and Robotic Surgery, Careggi University Hospital, Florence, Italy
| | - J Sven Mieog
- Department of Surgery, Leiden Universitair Medisch Centrum, Leiden, The Netherlands
| | - Daan J Lips
- Department of Surgery, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Olivier Saint-Marc
- Service de Chirurgie Digestive, Endocrinienne et Thoracique, Centre Hospitalier Universitaire Orleans, Orleans, France
| | - Misha D P Luyer
- Department of Surgery, Catharina Hospital, Eindhoven, The Netherlands
| | - Susan van Dieren
- Department of Surgery, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
| | - Geert Kazemier
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Felix Nickel
- Dept. of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | | | - Hjalmar C van Santvoort
- Department of Surgery, Regional Academic Cancer Centre Utrecht, St. Antonius Hospital and University Medical Centre, Utrecht, The Netherlands
| | | | - Roeland F de Wilde
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Mohammad Abu Hilal
- Department of Surgery, School of Medicine, University of Jordan, Amman, Jordan
- Department of Surgery, Southampton University, Southampton, UK
| | - Marc G Besselink
- Department of Surgery, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Zhang Y, Karimi SS, Requena DO, Khararjian A, Liang TZ, Mayer C, Birkness-Gartman J, Larman T, Montgomery EA, Voltaggio L. Intestinal and Liver Involvement by Endometrial Stromal Sarcoma: Expanding the Differential Diagnosis of Mesenchymal Tumors Involving the GI Tract. Hum Pathol 2025:105784. [PMID: 40319947 DOI: 10.1016/j.humpath.2025.105784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Endometrial stromal sarcoma (ESS) is a mesenchymal tumor known for infiltrative growth and lymphovascular invasion. Most examples arise in the endometrium, but some occur in the abdominal cavity without a primary uterine lesion, likely originating from endometriosis. ESS involving the gastrointestinal (GI) tract and liver is rare. We reviewed 15 examples from 13 women between 2010 and 2023. The mean age was 51 (range 16-81); just over half of the patients (7/13, 54%) had a history of uterine ESS. Twelve tumors involved the intestines and three the liver; rectosigmoid colon was the most affected site. Tumors averaged 4.6 cm (range 0.3-10 cm) and involved all bowel layers with two showing transmural involvement. Predominantly low-grade features were observed, including uniform round to spindle cells in fibrous or myxoid stroma, permeative growth, hyaline plaques, and spiral arterioles. Endometriosis was seen in 4 (27%) cases. Immunohistochemical analysis revealed positivity for ER (93%), PR (92%), CD10 (91%), and focal cyclin D1 (80%). A minority of cases showed staining with smooth muscle markers, CD117, DOG1, and keratins. High-grade features were suggested but not diagnostic in three cases. Follow-up data for three patients showed one death at 16 months, one patient disease-free at 94 months, and another alive at 16 months. ESS involving the GI tract is rare, may lack a uterine primary, and can mimic other neoplasms due to keratin, smooth muscle, and CD117/DOG1 expression. Recognizing ESS-associated features-such as a permeative pattern, hyaline plaques, and spiral arterioles-is key to avoiding misdiagnosis.
Collapse
Affiliation(s)
- Yujie Zhang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Saman S Karimi
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Domenika Ortiz Requena
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA.
| | | | - Tom Z Liang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Chen Mayer
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | | | - Tatianna Larman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Elizabeth Anne Montgomery
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA.
| | - Lysandra Voltaggio
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
9
|
Li J, Li Y, Fu L, Chen H, Du F, Wang Z, Zhang Y, Huang Y, Miao J, Xiao Y. Targeting ncRNAs to overcome metabolic reprogramming‑mediated drug resistance in cancer (Review). Int J Oncol 2025; 66:35. [PMID: 40116120 PMCID: PMC12002672 DOI: 10.3892/ijo.2025.5741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/07/2025] [Indexed: 03/23/2025] Open
Abstract
The emergence of resistance to antitumor drugs in cancer cells presents a notable obstacle in cancer therapy. Metabolic reprogramming is characterized by enhanced glycolysis, disrupted lipid metabolism, glutamine dependence and mitochondrial dysfunction. In addition to promoting tumor growth and metastasis, metabolic reprogramming mediates drug resistance through diverse molecular mechanisms, offering novel opportunities for therapeutic intervention. Non‑coding RNAs (ncRNAs), a diverse class of RNA molecules that lack protein‑coding function, represent a notable fraction of the human genome. Due to their distinct expression profiles and multifaceted roles in various cancers, ncRNAs have relevance in cancer pathophysiology. ncRNAs orchestrate metabolic abnormalities associated with drug resistance in cancer cells. The present review provides a comprehensive analysis of the mechanisms by which metabolic reprogramming drives drug resistance, with an emphasis on the regulatory roles of ncRNAs in glycolysis, lipid metabolism, mitochondrial dysfunction and glutamine metabolism. Furthermore, the present review aimed to discuss the potential of ncRNAs as biomarkers for predicting chemotherapy responses, as well as emerging strategies to target ncRNAs that modulate metabolism, particularly in the context of combination therapy with anti‑cancer drugs.
Collapse
Affiliation(s)
- Junxin Li
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yanyu Li
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Lin Fu
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Huiling Chen
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Fei Du
- Department of Pharmacy, The Fourth Affiliated Hospital of Southwest Medical University, Meishan, Sichuan 64200, P.R. China
| | - Zhongshu Wang
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yan Zhang
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yu Huang
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Jidong Miao
- Department of Oncology, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yi Xiao
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| |
Collapse
|
10
|
Dawood ZS, Khalil M, Waqar U, Banani I, Alidina Z, Pawlik TM. Use of textbook outcome as a quality metric in hepatopancreaticobiliary surgery: a systematic review and meta-analysis. J Gastrointest Surg 2025; 29:102005. [PMID: 40023393 DOI: 10.1016/j.gassur.2025.102005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Textbook outcomes (TOs) represent the optimal course after surgery. To date, no meta-analysis has assessed the pooled TOs of patients undergoing hepatopancreatobiliary (HPB) surgery and the effect of TO achievement on patient outcomes. This systematic review and meta-analysis aimed to assess TO achievement across different studies and to characterize the effect of TO achievement on patient-related outcomes, including disease-free survival (DFS) and overall survival (OS). METHODS PubMed, Embase, and Scopus databases were searched (1990-2024). The criteria used to define TO and the median overall TO in HPB surgery were obtained. In addition, a random-effects meta-analysis was conducted to assess the effect of TO achievement on 5-year OS and DFS. RESULTS A total of 27 studies involving 517,304 patients met inclusion criteria. The main criteria used to define TO included absence of readmission and mortality within 30 days after discharge, severe postoperative complications, prolonged hospital stay, and negative surgical margin (R0). Of note, the main factors related to TO achievement were younger patient age and lower American Society of Anesthesiologists score. Overall, the median rates of TOs achieved across procedures were 62.0% (IQR, 48.0%-69.0%) for hepatic procedure, 54.0% (IQR, 41.0%-68.0%) for biliary procedure, 46.0% (IQR, 42.0%-46.5%) for combined hepatopancreatic procedure, 45.0% (IQR, 30.5%-59.0%) for pancreatic procedure, 33.0% (IQR, 32.2%-34.0%) for liver transplantation, and 19.5% (IQR, 16.8%-22.3%) for combined hepatobiliary procedure. TO achievement was associated with improved odds of 5-year OS (odds ratio [OR], 1.22 [95% CI, 1.20-1.24]) and 5-year DFS (OR, 1.26 [95% CI, 1.16-1.37]). CONCLUSION Overall, hepatic and biliary operations had the highest TO achievement, followed by pancreatic procedures. In contrast, hepatobiliary surgery and liver transplantation had the lowest TO. There was a significant discrepancy in the definition of TO across different studies, highlighting the need for consensus on the definition of TO.
Collapse
Affiliation(s)
- Zaiba Shafik Dawood
- Department of Surgery, The Aga Khan University Hospital, Medical College, Aga Khan University, Karachi, Pakistan
| | - Mujtaba Khalil
- Department of Surgery, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Usama Waqar
- Department of Surgery, The Aga Khan University Hospital, Medical College, Aga Khan University, Karachi, Pakistan
| | - Illiyun Banani
- Department of Surgery, The Aga Khan University Hospital, Medical College, Aga Khan University, Karachi, Pakistan
| | - Zayan Alidina
- Department of Surgery, The Aga Khan University Hospital, Medical College, Aga Khan University, Karachi, Pakistan
| | - Timothy M Pawlik
- Department of Surgery, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States.
| |
Collapse
|
11
|
Shahbaz NK, Verhoeff K, Wees T, Jatana S, Quan D, Glinka J, Skaro A, Tang ES. Laparoscopic versus open pancreaticoduodenectomy outcomes in patients ≥ 75 years old: an NSQIP analysis of 4343 patients. HPB (Oxford) 2025; 27:696-705. [PMID: 39965982 DOI: 10.1016/j.hpb.2025.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/19/2025] [Accepted: 01/28/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND The benefits of MIS in older adults are conflicting. This study evaluates demographics and early outcomes, for older patients (≥75) undergoing minimally invasive (MIS) versus open pancreaticoduodenectomy (PD). METHOD We categorized elderly patients who underwent PD from 2017 to 2021 NSQIP databases by surgical approach (open vs MIS). Baseline characteristics were examined with bivariate analysis, and multivariate logistic regression assessed the independent effect of minimally invasive surgery on 30-day serious complications and mortality. RESULTS Amongst 4137 patients, 150 (3.63 %) underwent MIS PD. Patients demographics were similar. Open cohorts were older (79.1 vs 78.4 years; p = 0.011) with greater tumor invasion (36.6 % vs. 27.0 %; p = 0.018). MIS had longer operations (133.1 vs 119.6 min; p < 0.001). Multivariate analysis demonstrated that MIS approach was associated with increased serious complications (OR 2.21; p < 0.001), but not mortality (OR 2.11; p = 0.173). Post hoc analysis excluding cases converted to open demonstrated no difference in serious complications (OR 1.94; p = 0.070) or mortality (OR 3.58; p = 0.094). PSM analysis estimated a 14.7 % higher rate of serious complications in MIS but similar mortality (p = 0.291). CONCLUSIONS MIS PD uptake in elderly patients remains limited, with early findings indicating longer operations and higher complications. Further research on patient selection differences, technique modifications, and center expertise is required.
Collapse
Affiliation(s)
- Nazgol K Shahbaz
- Schulich School of Medicine and Dentistry, London, Ontario, Canada
| | - Kevin Verhoeff
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada.
| | - Tyrell Wees
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Sukhdeep Jatana
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | | | - Juan Glinka
- Department of Surgery, London, Ontario, Canada
| | - Anton Skaro
- Department of Surgery, London, Ontario, Canada
| | | |
Collapse
|
12
|
Chen Y, Xie T, Chen L, Zhang Z, Wang Y, Zhou Z, Liu W. The preoperative prediction of lymph node metastasis of resectable pancreatic ductal adenocarcinoma using dual-layer spectral computed tomography. Eur Radiol 2025; 35:2692-2701. [PMID: 39448418 DOI: 10.1007/s00330-024-11143-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/26/2024] [Accepted: 09/19/2024] [Indexed: 10/26/2024]
Abstract
OBJECTIVES To investigate the value of dual-layer spectral computed tomography (DLCT) parameters derived from primary tumors in predicting lymph node metastasis (LNM) of resectable pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS In this retrospective study, patients with resectable PDAC who underwent DLCT within 2-week intervals before surgery were enrolled and randomly divided into training and validation sets at a 7:3 ratio. The patients' clinical data, CT morphological features, and DLCT parameters were analyzed. Univariate and multivariate logistic analyses were used to identify the predictors and construct a predictive model, and receiver operator characteristic (ROC) curves were programmed to evaluate the predictive efficacy. RESULTS We enrolled 107 patients (44 patients with LNM and 63 patients without LNM). Among all variables, iodine concentration in the venous phase, extracellular volume, and tumor size were identified as independent predictors of LNM. The nomogram model, incorporating the two DLCT parameters and the morphological feature, achieved an area under the curve (AUC) of 0.877 (95% confidence interval [CI]: 0.803-0.952) and 0.842 (95% CI: 0.707-0.977) for predicting LNM in the training and validation sets, respectively. Furthermore, the AUC of the nomogram model was greater than that of morphological features of lymph nodes in the training (AUC = 0.877 vs. 0.570) and validation (AUC = 0.842 vs. 0.583) sets. CONCLUSIONS DLCT has the potential to predict LNM in patients with resectable PDAC and show a better predictive value than morphological features of lymph nodes. KEY POINTS Question Morphological features of lymph nodes are of limited value in detecting metastatic lymph nodes in pancreatic ductal adenocarcinoma (PDAC). Findings Dual-layer spectral computed tomography (DLCT) parameters and morphological features derived from PDAC lesions show good preoperatively predictive efficacy for lymph node metastasis. Clinical relevance The proposed DLCT-based nomogram model may serve as an effective and convenient tool for preoperatively predicting lymph node metastasis of resectable PDAC.
Collapse
Affiliation(s)
- Yi Chen
- Department of Radiology, Fudan University Shanghai Cancer Center & Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Tiansong Xie
- Department of Radiology, Fudan University Shanghai Cancer Center & Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Radiology, Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, 201100, China
| | - Lei Chen
- Department of Radiology, Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, 201100, China
| | - Zehua Zhang
- Department of Radiology, Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, 201100, China
| | - Yu Wang
- Clinical and Technical Support, Philips Healthcare, Shanghai, 200072, China
| | - Zhengrong Zhou
- Department of Radiology, Fudan University Shanghai Cancer Center & Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Department of Radiology, Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, 201100, China.
| | - Wei Liu
- Department of Radiology, Fudan University Shanghai Cancer Center & Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
13
|
Liu Y, Peng J, Zhao Y, Wang W. Emerging pathological diagnostic strategies for solid pseudopapillary neoplasm of the pancreas: insights from omics and innovative techniques. J Pathol Clin Res 2025; 11:e70029. [PMID: 40312910 PMCID: PMC12046068 DOI: 10.1002/2056-4538.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/21/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025]
Abstract
Solid pseudopapillary neoplasm (SPN) of the pancreas is a rare, low-grade malignant tumor, representing 0.9-2.7% of all exocrine pancreatic tumors. SPN patients generally have a favorable prognosis with a 5-year survival rate exceeding 95% following complete surgical resection. Accurate diagnosis is crucial to avoid unnecessary treatments. Currently, SPN diagnosis relies on imaging techniques such as CT and MRI, along with immunohistochemical analysis of biopsy and resection samples. The main challenge in diagnosis is the potential inability to accurately identify recurrent or metastatic SPN, as well as 'malignant' SPN, due to the lack of specific biomarkers. Advances in high-throughput omics technologies, including genomics, transcriptomics, proteomics and metabolomics, have opened new avenues for identifying novel biomarkers for SPN. Additional, liquid biopsy techniques have enabled more comprehensive analysis of biosamples such as pancreatic cyst fluid, offering promising prospects for preoperative diagnosis. This review highlights recent research on SPN diagnosis, focusing on immunohistochemical markers, tissue sampling methods and the potential of omics approaches. It also discusses the challenges and opportunities in improving diagnostic accuracy, particularly for high-grade and metastatic SPNs.
Collapse
Affiliation(s)
- Yuanhao Liu
- Department of PathologyPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
| | - Junya Peng
- Institute of Clinical MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
- State Key Laboratory of Complex, Severe, and Rare DiseasesPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
| | - Yupei Zhao
- State Key Laboratory of Complex, Severe, and Rare DiseasesPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
- Department of General SurgeryPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
- Department of Basic Medical SciencesSchool of Medicine, Tsinghua UniversityBeijingPR China
- Peking University‐Tsinghua Center for Life SciencesBeijingPR China
| | - Wenze Wang
- Department of PathologyPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
- Molecular Pathology Research Center, Department of PathologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPR China
| |
Collapse
|
14
|
Ling D, Xiang C, Guolin H, Huisheng S, Xiaohua N. Ellipticine targets FGFR3 to mediate the RAS/MAPK-P38 signalling pathway to induce apoptosis in hepatocellular carcinoma cells. 3 Biotech 2025; 15:111. [PMID: 40191451 PMCID: PMC11968639 DOI: 10.1007/s13205-025-04269-7] [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: 01/07/2025] [Accepted: 03/12/2025] [Indexed: 04/09/2025] Open
Abstract
This study aimed to investigate the toxic effects of ellipticine on liver cancer cells and predict its anti-liver cancer mechanism through network pharmacology, especially by targeting FGFR3 to regulate the RAS/MAPK-P38 signaling pathway, thereby inducing apoptosis of liver cancer cells. The inhibitory effect of ellipticine on the proliferation of HepG2, Huh-7, SMMC7721, BEL-7402, SK-HEP-1, LX-2, and MHCC97H cells was detected by CCK-8 assay, and the IC50 value was calculated. The potential targets of ellipticine were predicted by the database, and the intersection analysis with liver cancer-related targets was performed to construct a protein interaction network (PPI), (KEGG) pathway enrichment analysis, and molecular docking verification. FGFR3 in HepG2 cells was knocked down by siRNA, and the effects on cell proliferation, apoptosis, and ROS levels were observed. The expression changes of FGFR3, RAS, P38, and their phosphorylated forms after ellipticine treatment, as well as the effects of RAS agonist ML-908 and P38 inhibitor PD169316 on cell proliferation, apoptosis, and migration, were detected by Western blotting. Ellipticine has an inhibitory effect on all tested liver cancer cell lines, among which HepG2 has the strongest inhibitory effect, with an IC50 of 5.15 ± 0.25 μM. Ellipticine is predicted to have 32 potential targets, and 5 common targets among the 225 targets related to liver cancer, including PDGFRA, KIT, FGFR3, ERBB2, and STAT3. KEGG analysis showed that these targets are mainly involved in cancer pathways. Molecular docking showed that Ellipticine can bind strongly to FGFR3. FGFR3 expression is highest in HepG2 cells. After knocking down FGFR3, the proliferation ability of HepG2 cells is further weakened, and the addition of apoptosis inhibitor ZVAD can partially restore the proliferation ability. ROS levels increase after Ellipticine treatment, and ROS levels further increase after knocking down FGFR3, and ZVAD treatment can reduce ROS levels. After Ellipticine treatment, the expression levels of FGFR3, RAS, and p-P38 decrease. Ellipticine-induced cell proliferation inhibition and apoptosis were reversed by RAS agonist ML-908, whereas P38 inhibitor PD169316 exacerbated cell apoptosis and migration inhibition. Ellipticine induces apoptosis of liver cancer cells by targeting FGFR3 and inhibiting the RAS/MAPK-P38 signaling pathway. This discovery provides new mechanistic insights into Ellipticine as a liver cancer treatment and may lay the foundation for the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Deng Ling
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Guangzhou, China
| | - Chen Xiang
- Department of General Surgery, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Guangzhou, China
| | - Hu Guolin
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Guangzhou, China
| | - Song Huisheng
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Guangzhou, China
| | - Niu Xiaohua
- Department of General Surgery, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
15
|
Fall S, Souche R, Bardol T, Fabre JM, Borie F. Laparoscopic versus open pancreaticoduodenectomy for pancreatic or periampullary tumors: a multicenter propensity score-matched comparative study. Surg Endosc 2025; 39:3037-3048. [PMID: 40131468 DOI: 10.1007/s00464-025-11677-6] [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: 12/29/2024] [Accepted: 03/14/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Pancreaticoduodenectomy (PD) is a complex surgical procedure for which the use of minimally invasive approaches is still debated. The present study aims to compare the outcomes after laparoscopic pancreaticoduodenectomy (LPD) versus open pancreaticoduodenectomy (OPD) for pancreatic head and peri-ampullary tumors. METHODS This is a retrospective multicenter study comparing 132 patients who underwent LPD with a historic cohort of OPD performed before LPD program implementation, by two pancreatic and laparoscopic surgeons in tertiary centers. After propensity score-matching (PSM), 85 LPD patients were matched with 85 OPD patients with similar preoperative characteristics and 90-day outcomes were compared. RESULTS LPD was associated with significantly longer operative time (355 vs. 269 min; P = 0.001). Estimated blood loss were not different (median = 211 vs 277 mL; P = 0.141) and the conversion to open rate for LPD was 8.33% in the original cohort and 6% after PSM. LPD was not statistically different from OPD for severe complications rates (17.6% vs 24.7%; P = 0.348), clinically relevant POPF (17.6% vs 25.8%; P = 0.265), biliary leakage (17.6% vs 25.8%; P = 0.265), hemorrhage (12.9% vs 14.1%; P = 0.999), delayed gastric emptying rate (10.5% vs 5.8%; P = 0.404) and reoperation rate (12.9% vs 11.7%; P = 0.999). Length of stay in the LPD group was shorter than in the OPD group (15.13 vs. 19.44 days; P = 0.01). Mortality rates (3.5% vs. 0%; P = 0.245) did not differ between LPD and OPD groups. Regarding final histology, we found a higher number of lymph nodes in the LPD group (15.48 vs 13.46; P = 0.017) without difference on R0 rates. CONCLUSION LPD is a safe procedure when performed by pancreatic and laparoscopic surgeons and could lead to a shorter hospital stay and improve lymphadenectomy with similar postoperative outcomes to OPD.
Collapse
Affiliation(s)
- Seïla Fall
- Oncologic and Minimally Invasive Digestive Surgery Unit, Department of Digestive Surgery and Transplantation, Saint Eloi Hospital, University of Montpellier - Nîmes, 80 Avenue Augustin Fliche, 34295, Montpellier, France
- Digestive Surgery Department, Carémeau Hospital, University of Montpellier - Nîmes, Place du Professeur Debré, 30900, Nîmes, France
| | - Regis Souche
- Oncologic and Minimally Invasive Digestive Surgery Unit, Department of Digestive Surgery and Transplantation, Saint Eloi Hospital, University of Montpellier - Nîmes, 80 Avenue Augustin Fliche, 34295, Montpellier, France.
| | - Thomas Bardol
- Oncologic and Minimally Invasive Digestive Surgery Unit, Department of Digestive Surgery and Transplantation, Saint Eloi Hospital, University of Montpellier - Nîmes, 80 Avenue Augustin Fliche, 34295, Montpellier, France
| | - Jean-Michel Fabre
- Oncologic and Minimally Invasive Digestive Surgery Unit, Department of Digestive Surgery and Transplantation, Saint Eloi Hospital, University of Montpellier - Nîmes, 80 Avenue Augustin Fliche, 34295, Montpellier, France
| | - Frederic Borie
- Digestive Surgery Department, Carémeau Hospital, University of Montpellier - Nîmes, Place du Professeur Debré, 30900, Nîmes, France
| |
Collapse
|
16
|
Geng Y, Luo K, Stam J, Oosterhuis D, Gorter AR, van den Heuvel M, Crescitelli R, de Meijer VE, Wolters JC, Olinga P. Characterization of Extracellular Vesicles Derived From Human Precision-Cut Liver Slices in Metabolic Dysfunction-Associated Steatotic Liver Disease. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70043. [PMID: 40313415 PMCID: PMC12042696 DOI: 10.1002/jex2.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/30/2025] [Accepted: 02/28/2025] [Indexed: 05/03/2025]
Abstract
Extracellular vesicles (EVs) are cell-produced, membrane-surrounded vesicles that harbour the biological features of donor cells. In the current study, we are the first to isolate and characterize EVs isolated from human precision-cut liver slices (PCLS), obtained from both healthy and metabolic dysfunction-associated steatohepatitis (MASH) cirrhotic livers. PCLS derived from patients can faithfully represent disease conditions in humans. EVs were isolated from human PCLS after incubating in normal medium or modified medium that mimics the pathophysiological environment of metabolic dysfunction associated liver disease (MASLD). MASH PCLS produced higher amounts of EVs compared to healthy PCLS (p < 0.001). Mass spectrometry revealed that around 300 proteins were significantly different in EVs derived from MASH PCLS versus healthy PCLS (FDR < 0.05), irrespective of the type of medium. Significantly changed EV proteins were largely involved in signalling receptor binding function and showed potential in promoting fibrosis. In the liver, these ligand-associated receptors are highly expressed in hepatic stellate cells, and the MASH EVs functionally promoted the activation of hepatic stellate cells. Furthermore, the amounts of EpCAM and ITGA3 in EVs were positively associated with the progression of MASLD, which suggests the use of liver-derived EVs as potential biomarkers for MASLD. Characterization of EVs derived from human PCLS may assist future studies in investigating the pathogenesis and identifying liver-specific EVs as biomarkers of MASLD.
Collapse
Affiliation(s)
- Yana Geng
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of PharmacyUniversity of GroningenGroningenthe Netherlands
| | - Ke Luo
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of PharmacyUniversity of GroningenGroningenthe Netherlands
| | - Janine Stam
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of PharmacyUniversity of GroningenGroningenthe Netherlands
- Department of Analytical Biochemistry, Groningen Research Institute of PharmacyUniversity of GroningenGroningenthe Netherlands
| | - Dorenda Oosterhuis
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of PharmacyUniversity of GroningenGroningenthe Netherlands
| | - Alan R. Gorter
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of PharmacyUniversity of GroningenGroningenthe Netherlands
| | - Marius van den Heuvel
- Division of Pathology, Department of Pathology and Medical BiologyUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
| | - Rossella Crescitelli
- Department of Surgery, Sahlgrenska Center for Cancer Research and Wallenberg Centre for Molecular and Translational Medicine, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGöteborgSweden
| | - Vincent E. de Meijer
- Department of Surgery, Section of Hepatobiliary Surgery & Liver TransplantationUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
| | - Justina C. Wolters
- Department of PediatricsUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of PharmacyUniversity of GroningenGroningenthe Netherlands
| |
Collapse
|
17
|
Zhang J, Lin F, Xu Y, Sun J, Zhang L, Chen W. Lactylation and Ischemic Stroke: Research Progress and Potential Relationship. Mol Neurobiol 2025; 62:5359-5376. [PMID: 39541071 DOI: 10.1007/s12035-024-04624-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Ischemic stroke is caused by interrupted cerebral blood flow and is a leading cause of mortality and disability worldwide. Significant advancements have been achieved in comprehending the pathophysiology of stroke and the fundamental mechanisms responsible for ischemic damage. Lactylation, as a newly discovered post-translational modification, has been reported to participate in several physiological and pathological processes. However, research on lactylation and ischemic stroke is scarce. This review summarized the current function of protein lactylation in other diseases or normal physiological processes and explored their potential link with the pathophysiological process and the reparative mechanism of ischemic stroke. We proposed that neuroinflammation, regulation of metabolism, regulation of messenger RNA translation, angiogenesis, and neurogenesis might be the bridge linking lactylation and ischemic stroke. Our study provided a novel perspective for comprehending the role of protein lactylation in the pathophysiological processes underlying ischemic stroke. Lactylation might be a promising target in drug development of ischemic stroke.
Collapse
Affiliation(s)
- Jingyuan Zhang
- Department of Cerebrovascular Disease, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Feng Lin
- Department of Cerebrovascular Disease, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Yue Xu
- Department of Cerebrovascular Disease, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Jiaxin Sun
- Department of Cerebrovascular Disease, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Lei Zhang
- Department of Cerebrovascular Disease, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China.
| | - Wenli Chen
- Department of Pharmacy, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China.
| |
Collapse
|
18
|
Yong GY, Kamkaew A, Kue CS. Synergistic approach of PEGylated photothermal agent and immunomodulator in cancer immunotherapy. Nanomedicine (Lond) 2025; 20:967-983. [PMID: 40214079 PMCID: PMC12051527 DOI: 10.1080/17435889.2025.2489342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 04/02/2025] [Indexed: 05/02/2025] Open
Abstract
Photothermal therapy (PTT) utilizes photothermal agents (PTAs) to generate heat at the local tumor site that leads to ablation upon photoirradiation at a specific wavelength of light. Currently, most of the available PTAs have weak tumor selectivity and depositing ability, which leads to poor therapeutic outcomes. PEGylation of PTAs improves therapeutic outcomes, prolongs systemic circulation time, enhances tumor accumulation, and reduces the risk of clearance by the immune system. This paper reviews the recent developments of PEGylated PTAs in photothermal cancer therapy from 2019 to 2023, highlighting their antitumour efficacy and immune response post-therapy with immune agents, current challenges and strategies. This review aims to foster knowledge dissemination on the application of nanomedicine in photothermal cancer therapy from an immunological perspective and to encourage the clinical translation of these nanomaterials.
Collapse
Affiliation(s)
- Gong Yi Yong
- School of Graduate Studies, Management and Science University, Shah Alam, Malaysia
| | - Anyanee Kamkaew
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Chin Siang Kue
- Faculty of Health and Life Sciences, Management and Science University, Shah Alam, Malaysia
| |
Collapse
|
19
|
Chen H, Zhao W, Xiao Y, Gao Q, Yang X, Pang K, Huang B, Liang X. Association between dietary niacin intake and the odds of gallstones in US adults: A cross-sectional study in NHANES 2017-2020. Prev Med Rep 2025; 53:103057. [PMID: 40264748 PMCID: PMC12013329 DOI: 10.1016/j.pmedr.2025.103057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/24/2025] Open
Abstract
Objective To investigate the association between dietary niacin intake and the risk of gallstones in American adults using data from the National Health and Nutrition Examination Survey (NHANES) from 2017 to 2020. Methods This cross-sectional study analyzed data from 8191 participants aged 18 years and older. Dietary niacin intake was assessed using two 24-h dietary recalls. The presence of gallstones was identified through a questionnaire. Logistic regression models were used to estimate odds ratios (ORs) and 95 % confidence intervals (CIs) for gallstones across quartiles of niacin intake, adjusting for demographic and health-related covariates. Results Participants with higher niacin intake showed a significantly lower risk of gallstones. After adjusting for a wide range of covariates, individuals in the highest quartile of niacin intake had a 49 % reduced risk of gallstones compared to those in the lowest quartile (OR = 0.51, 95 % CI: 0.34, 0.76). Conclusion Higher dietary niacin intake is associated with a reduced risk of gallstones in US adults. These findings suggest that increasing niacin intake could be a viable strategy for the prevention of gallstones. Future longitudinal studies are needed to confirm these results and explore the underlying mechanisms.
Collapse
Affiliation(s)
- Huadi Chen
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, People's Republic of China
| | - Wenting Zhao
- Development Planning Department, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, People's Republic of China
| | - Yi Xiao
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, People's Republic of China
| | - Qiaoping Gao
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, People's Republic of China
| | - Xiaoqu Yang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, People's Republic of China
| | - Kangfeng Pang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, People's Republic of China
| | - Baoyi Huang
- Department of Breast Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, People's Republic of China
| | - Xiaolu Liang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, People's Republic of China
| |
Collapse
|
20
|
Ricci C, D'Ambra V, Alberici L, Ingaldi C, Minghetti M, Bonini G, Casadei R. Minimal Invasive Pancreatoduodenectomy: A Comprehensive Systematic Review and Metanalysis of Randomized Controlled Clinical Trials. Ann Surg Oncol 2025; 32:3614-3622. [PMID: 39937403 PMCID: PMC11976793 DOI: 10.1245/s10434-025-16990-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: 11/14/2024] [Accepted: 01/23/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND The role of a minimally invasive approach (MI) in patients who underwent pancreatoduodenectomy (PD) remained unclear. METHODS A systematic search of randomized controlled trials was conducted. A random-effects meta-analysis was conducted, reporting risk ratio (RR) or mean difference (MD). The primary endpoints were the morbidity, mortality, and R1 rate. The secondary endpoints were clinically relevant postoperative pancreatic fistula (POPF), postpancreatectomy hemorrhage (PPH), delayed gastric emptying (DGE), biliary fistula, reoperation, length of stay (LOS), time to functional recovery (TFR), and readmission. RESULTS The meta-analysis includes seven studies and 1428 patients: 618 (46.5%) in the OPD arm and 711 (53.5%) in minimally invasive pancreaticoduodenectomy (MIPD). The mortality rate was 2.9% for MIPD and 2.6% for OPD (RR 1.11 [range 0.53-2.29]). The major morbidity rate was 29.4% for MIPD and 25.6% for OPD (RR 1.11 [range 0.53-2.29]). The R1 rate was 6.2% for MIPD and 7% for OPD (RR 0.80 [0.54-1.20]). The operative time, comprehensive complication index score, POPF, PPH, DGE, biliary fistula, reoperation, readmission, LOS, TFR, and harvested lymph nodes were similar. Greater than 25% of heterogeneity was observed for major morbidity, operative time, POPF, LOS, TFR, and harvested lymph nodes. No publication bias was registered. CONCLUSIONS Minimally invasive pancreaticoduodenectomy was not superior to OPD and provided marginal advantages in short-term results. Further efforts should be addressed to clarify the impact of learning curve in MIPD results and the economic sustainability of MIPD, particularly robotic approach.
Collapse
Affiliation(s)
- Claudio Ricci
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, Bologna, Italy.
| | - Vincenzo D'Ambra
- Division of Pancreatic Surgery, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Laura Alberici
- Division of Pancreatic Surgery, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Carlo Ingaldi
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Margherita Minghetti
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Division of Pancreatic Surgery, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Giulia Bonini
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Division of Pancreatic Surgery, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Riccardo Casadei
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Division of Pancreatic Surgery, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| |
Collapse
|
21
|
Vijayan V, M Unagolla J, Panchal D, John JE, Menon SS, Menon JU. Biomimetic nanoparticles for targeted therapy of liver disease. RSC PHARMACEUTICS 2025:d5pm00044k. [PMID: 40321406 PMCID: PMC12045541 DOI: 10.1039/d5pm00044k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/25/2025] [Indexed: 05/08/2025]
Abstract
Liver fibrosis is a progressive and fatal condition characterized by stiffness and scarring of the liver due to excessive buildup of extracellular matrix (ECM) proteins. If left untreated, it can progress to liver cirrhosis and hepatocellular carcinoma (HCC)-one of the fastest-rising causes of cancer mortality in the United States. Despite the increased prevalence of liver fibrosis due to infections, exposure to toxins, and unhealthy lifestyles, there are no effective treatments available. Recent advances in nanomedicine can lead to more targeted and effective strategies for treating liver diseases than existing treatments. In particular, the use of biomimetic nanoparticles (NPs) such as liposomes and cell-membrane-coated NPs is of interest. NPs functionalized with cell membranes mimic the properties of the source cell used and provide inherent immune evasion ability, homologous adhesion, and prolonged circulation. This review explores the types of biomimetic coatings, different cargoes delivered through biomimetic NPs for various treatment modalities, and the type of core NPs used for targeting liver fibrosis and HCC.
Collapse
Affiliation(s)
- Veena Vijayan
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Kingston RI 02881 USA
| | - Janitha M Unagolla
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Kingston RI 02881 USA
| | - Dhruvisha Panchal
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Kingston RI 02881 USA
| | - Judith Eloyi John
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Kingston RI 02881 USA
| | | | - Jyothi U Menon
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Kingston RI 02881 USA
- Department of Chemical Engineering, University of Rhode Island Kingston RI 02881 USA
| |
Collapse
|
22
|
Wang Y, Ma X, Ma J, Li J, Lin Z, Gao W, Gong P, Dai P. Thyroid dysfunction as a predictor of PD- 1/PD-L1 inhibitor efficacy in advanced lung cancer. BMC Cancer 2025; 25:791. [PMID: 40295950 PMCID: PMC12036238 DOI: 10.1186/s12885-025-14097-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
PURPOSE To investigate the correlation between thyroid dysfunction (TD) and the efficacy of programmed cell death protein 1 (PD-1) and programmed death ligand 1 (PD-L1) inhibitors in the treatment of advanced lung cancer, and the possible influencing factors for TD occurrence, providing insights that could guide individualized therapeutic approaches. METHODS The data of 120 advanced lung cancer patients from January 2019 to August 2024 were retrospectively collected. Then, the patients were divided into TD and non-TD subgroups according to whether TD occurred or not, to analyse the possible factors influencing the occurrence of TD and the correlation between TD and PD-1/PD-L1 inhibitor efficacy. RESULTS For all cases, the baseline TSH level was significantly higher in the TD subgroup than in the non-TD subgroup (median: 2.33 mIU/L vs. 1.58 mIU/L, p = 0.001). The progression-free survival (PFS) was significantly longer in the TD subgroup than in the non-TD subgroup (mPFS: 7.90 months vs. 4.87 months, p = 0.003), and the patients in the TD subgroup had a lower HR for progression (0.499, 95% CI (0.317-0.766)). For the PD-1/PD-L1 inhibitor group, the baseline TSH level was also significantly higher in the TD subgroup than in the non-TD subgroup (median: 2.16 mIU/L vs. 1.52 mIU/L, p = 0.009). The PFS was also significantly longer in the TD subgroup than in the non-TD subgroup (mPFS: 8.83 months vs. 6.50 months, p = 0.041). CONCLUSIONS The baseline TSH level was the predictive factor for the occurrence of TD. The occurrence of TD was positively associated with a favorable prognosis for patients with advanced lung cancer.
Collapse
Affiliation(s)
- Yanling Wang
- Department of Oncology, The First Affiliated Hospital of School of Medicine Shihezi University, Shihezi, Xinjiang, 832000, People's Republic of China
| | - Xiaoping Ma
- Department of Oncology, The First Affiliated Hospital of School of Medicine Shihezi University, Shihezi, Xinjiang, 832000, People's Republic of China
| | - Jia Ma
- Department of General Surgery, Shanghai Jian Gong Hospital, Shanghai, 200083, People's Republic of China
| | - Jing Li
- Department of Oncology, The First Affiliated Hospital of School of Medicine Shihezi University, Shihezi, Xinjiang, 832000, People's Republic of China
| | - Zhiyi Lin
- Department of Oncology, The First Affiliated Hospital of School of Medicine Shihezi University, Shihezi, Xinjiang, 832000, People's Republic of China
| | - Wei Gao
- Department of Radiotherapy, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, People's Republic of China.
| | - Ping Gong
- School of Medicine, Shihezi University, Shihezi, Xinjiang, 832000, People's Republic of China.
| | - Ping Dai
- Department of Radiotherapy, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, People's Republic of China.
| |
Collapse
|
23
|
Hu R, Tran B, Li S, Stackpole ML, Zeng W, Zhou Y, Melehy A, Sadeghi S, Finn RS, Zhou XJ, Li W, Agopian VG. Noninvasive prognostication of hepatocellular carcinoma based on cell-free DNA methylation. PLoS One 2025; 20:e0321736. [PMID: 40279344 PMCID: PMC12026916 DOI: 10.1371/journal.pone.0321736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 03/11/2025] [Indexed: 04/27/2025] Open
Abstract
BACKGROUND The current noninvasive prognostic evaluation methods for hepatocellular carcinoma (HCC), which are largely reliant on radiographic imaging features and serum biomarkers such as alpha-fetoprotein (AFP), have limited effectiveness in discriminating patient outcomes. Identification of new prognostic biomarkers is a critical unmet need to improve treatment decision-making. Epigenetic changes in cell-free DNA (cfDNA) have shown promise in early cancer diagnosis and prognosis. Thus, we aim to evaluate the potential of cfDNA methylation as a noninvasive predictor for prognostication in patients with active, radiographically viable HCC. METHODS Using Illumina HumanMethylation450 array data of 377 HCC tumors and 50 adjacent normal tissues obtained from The Cancer Genome Atlas (TCGA), we identified 158 HCC-related DNA methylation markers associated with overall survival (OS). This signature was further validated in 29 HCC tumor tissue samples. Subsequently, we applied the signature to an independent cohort of 52 patients with plasma cfDNA samples by calculating the cfDNA methylation-based risk score (methRisk) via random survival forest models with 10-fold cross-validation for the prognostication of OS. RESULTS The cfDNA-based methRisk showed strong discriminatory power when evaluated as a single predictor for OS (3-year AUC = 0.81, 95% CI: 0.68-0.94). Integrating the methRisk with existing risk indices like Barcelona clinic liver cancer (BCLC) staging significantly improved the noninvasive prognostic assessments for OS (3-year AUC = 0.91, 95% CI: 0.80-1), and methRisk remained an independent predictor of survival in the multivariate Cox model (P = 0.007). CONCLUSIONS Our study serves as a pilot study demonstrating that cfDNA methylation biomarkers assessed from a peripheral blood draw can stratify HCC patients into clinically meaningful risk groups. These findings indicate that cfDNA methylation is a promising noninvasive prognostic biomarker for HCC, providing a proof-of-concept for its potential clinical utility and laying the groundwork for broader applications.
Collapse
Affiliation(s)
- Ran Hu
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
- Bioinformatics Interdepartmental Graduate Program, University of California at Los Angeles, Los Angeles, California, United States of America
- Institute for Quantitative and Computational Biosciences, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Benjamin Tran
- Department of Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Shuo Li
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Mary L. Stackpole
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Weihua Zeng
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Yonggang Zhou
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Andrew Melehy
- Department of Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Saeed Sadeghi
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Richard S. Finn
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Xianghong Jasmine Zhou
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
- Institute for Quantitative and Computational Biosciences, University of California at Los Angeles, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Wenyuan Li
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Vatche G. Agopian
- Department of Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
24
|
Zhang L, Zhang S, Yan Y, Su C, Gao L, Li F, Li J, Gai Y, Zhang G, Zhang D. Diagnostic utility of ultrasonography in the management of postoperative fluid collections and abdominal indwelling catheters following pancreaticoduodenectomy: retrospective cohort study. Eur J Med Res 2025; 30:319. [PMID: 40270062 PMCID: PMC12016473 DOI: 10.1186/s40001-025-02590-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 04/13/2025] [Indexed: 04/25/2025] Open
Abstract
INTRODUCTION The management of postoperative fluid collections, which refers to the accumulation of fluid in the peritoneal cavity following pancreaticoduodenectomy, presents significant challenges. However, ultrasonography has emerged as a promising tool for diagnosing and guiding interventions for this condition. Ultrasonography offers several advantages, including accessibility, cost-effectiveness, and real-time imaging capabilities. It plays a crucial role in identifying ascitic fluid collections, characterizing their contents, and evaluating the severity of fluid collections. Moreover, ultrasound guidance enhances the safety and effectiveness of placing abdominal catheters. The aim of this study is to assess the diagnostic utility of ultrasonography in postoperative fluid collections following pancreaticoduodenectomy and evaluate the clinical efficacy of ultrasound-guided abdominal catheter placement. METHODS A total of 309 hospitalized patients underwent postoperative pancreaticoduodenectomy, with 171 patients undergoing laparoscopic pancreaticoduodenectomy (LPD) and 138 patients undergoing open pancreaticoduodenectomy (OPD), as assessed by ultrasonography. We examined the abdominal cavity for the presence of postoperative fluid collections and evaluated the site of postoperative fluid collections and the necessity for tube drainage. In cases where an abdominal indwelling catheter was required, we observed the location of postoperative fluid collections, performed echocardiography, and analyzed the characteristics of drainage fluid. We conducted a comparative analysis of short-term postoperative outcomes between LPD and OPD, encompassing hospitalization duration, fever duration, presence or localization of postoperative fluid collections, number of abdominal indwelling catheters used, location of abdominal drainage fluid collection, and time until postoperative catheter removal. RESULTS The LPD group demonstrated a significantly lower incidence of postoperative fluid collections compared to the OPD group, as determined by ultrasonography (39.2% vs. 59.3%, p = 0.001). Additionally, the LPD group had shorter hospital stays (16 [13, 21] vs. 21 [17, 28] days; p < 0.001), reduced duration of fever (1 [0, 3] vs. 3 [1, 5] days; p < 0.001), faster time to postoperative catheterization (7 [5, 10] vs. 8 [6, 13] days; p < 0.001), fewer required tubes (0 [0, 1] vs. 1 [0, 1]; p < 0.001), and shorter extubation time (7 [5, 9] vs. 9 [5, 12] h; p < 0.001) compared to the OPD group. There were correlations observed between the two groups regarding postoperative fluid collections, ultrasound sound transmission, separation of postoperative fluid collections, and traits of drainage fluid. However, there were no significant differences between the two groups in terms of postoperative fluid collections location (dissociative or restrictive), ultrasound sound transmission (excellent or poor), and separation of postoperative fluid collections (no separation, less separation, and more separation). CONCLUSIONS Postoperative fluid collections is a commonly encountered concurrent condition following pancreaticoduodenectomy. Ultrasonography allows for the observation of diverse characteristics related to postoperative fluid collections, including its precise localization, sound transmission properties, and the presence of internal separations. Moreover, it enables timely guidance for precise placement of drainage tubes.
Collapse
Affiliation(s)
- Lingyun Zhang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Suzhen Zhang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ye Yan
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Chen Su
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Li Gao
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Feng Li
- Department of Pancreatic Surgery, General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Jianzhi Li
- Department of Ultrasound, Shandong Public Health Clinical Center, Jinan, China
| | - Yonghao Gai
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guoquan Zhang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Dawei Zhang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China.
| |
Collapse
|
25
|
Wang X, Duan W, Ma Z, Wen H, Mao X, Liu C. ETV4/ALYREF-mediated glycolytic metabolism through PKM2 enhances resistance to ferroptosis and promotes the development of intrahepatic cholangiocarcinoma. Cancer Metab 2025; 13:19. [PMID: 40264211 PMCID: PMC12013154 DOI: 10.1186/s40170-025-00387-1] [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: 10/12/2024] [Accepted: 04/07/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (ICC) is the second most common primary hepatocellular cancer. This study investigated whether ETV4, ALYREF, and PKM2 affect glycolytic metabolism and ferroptosis, thereby potentially influencing ICC. METHODS Bioinformatic analysis was used to explore the expression levels and prognosis of ETV4, ALYREF, and PKM2 in ICC and their regulatory relationships were confirmed using in vitro experiments. Glycolytic metabolism and ferroptosis were examined, and chromatin immunoprecipitation and RNA immunoprecipitation experiments were performed to verify whether the ETV4, PKM2, and ALYREF could bind. The effect of ETV4/ALYREF on ICC was further confirmed by in vivo experiments. RESULTS ETV4, ALYREF, and PKM2 were highly expressed in ICC. Overexpressed (oe)-ETV4 and oe-PKM2 promoted cell migration and increased glucose (GLU) utilization and lactate and intracellular adenosine triphosphate (ATP) production. Addition of the ferroptosis inducer Erastin to the above groups revealed that sh-ETV4 and sh-ALYREF increased lipid reactive oxygen species (ROS), malondialdehyde (MDA), and Fe2+ levels, and oe-PKM2 reversed these effects in the sh-ETV4 and sh-ALYREF groups. Oe-ETV4 promoted the expression of PKM2, whereas sh-ALYREF inhibited the same. ETV4 could bind to ALYREF and PKM2 promoter, and ALYREF could promote the stability of PKM2 in an m5C-dependent manner. In vivo, ETV4 promotes tumor growth and the expression of proteins related to glycolytic metabolism by regulating ALYREF. CONCLUSION ETV4 promotes ICC development and ferroptosis resistance by facilitating glycolytic metabolism, and regulating PKM2 transcription by directly binding to the PKM2 promoter. Additionally, it mediates m5C-dependent PKM2 stabilization by directly binding to ALYREF. This study identified a new potential therapeutic target for ICC.
Collapse
Affiliation(s)
- Xiaohui Wang
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No. 61, Jiefang West Road, Furong District, Changsha, 410000, Hunan, China
| | - Wenbin Duan
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No. 61, Jiefang West Road, Furong District, Changsha, 410000, Hunan, China
| | - Zhongzhi Ma
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No. 61, Jiefang West Road, Furong District, Changsha, 410000, Hunan, China
| | - Haoquan Wen
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No. 61, Jiefang West Road, Furong District, Changsha, 410000, Hunan, China
| | - Xianhai Mao
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No. 61, Jiefang West Road, Furong District, Changsha, 410000, Hunan, China
- Department of Hepatobiliary and Intestinal Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410013, Changsha, China
| | - Changjun Liu
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No. 61, Jiefang West Road, Furong District, Changsha, 410000, Hunan, China.
| |
Collapse
|
26
|
Zhu Y, Yang Y, Lan Y, Yang Z, Gao X, Zhou J. The role of PKM2-mediated metabolic reprogramming in the osteogenic differentiation of BMSCs under diabetic periodontitis conditions. Stem Cell Res Ther 2025; 16:186. [PMID: 40251642 PMCID: PMC12008901 DOI: 10.1186/s13287-025-04301-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/01/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Diabetes mellitus (DM) and periodontitis have a bidirectional relationship, with each being a high-risk factor for the other. Prolonged hyperglycemia exacerbates periodontal inflammation and disrupts bone homeostasis. Pyruvate kinase M2 (PKM2), a key enzyme in glycolysis, is involved in metabolic reprogramming, but its role in osteogenesis under high-glucose (HG) inflammatory conditions remains largely unknown. This study aimed to investigate the effects of HG and inflammation on bone marrow mesenchymal stem cells (BMSCs) under indirect co-culture conditions and to explore how PKM2 regulates metabolism and mitochondrial function during osteogenic differentiation in HG inflammatory environments, elucidating its role in diabetic periodontitis (DP). METHODS Expose BMSCs to conditioned medium (CM) collected from RAW264.7 cells stimulated with HG and/or lipopolysaccharide (LPS). BMSCs functionality was assessed using CCK8, EdU, Annexin V-PI apoptosis assay, alkaline phosphatase (ALP), and Alizarin Red S (ARS) staining. Metabolic characteristics were evaluated through Seahorse assays, lactate production, glucose uptake, and ATP measurements. Mitochondrial function was assessed via JC-1, and ROS staining, Mito-Tracker staining, and transmission electron microscopy (TEM). Gene and protein expression were analyzed by quantitative real-time PCR and western blotting. In vivo therapeutic effects of shikonin were validated via micro-CT and histological staining in a diabetic periodontitis mouse model. RESULTS In vitro experiments demonstrated that HG inflammatory conditions impaired the survival of BMSCs, suppressed osteogenic differentiation, and induced metabolic reprogramming. This reprogramming was characterized by enhanced glycolysis, impaired oxidative phosphorylation (OXPHOS), abnormal upregulation of PKM2 expression, and mitochondrial dysfunction accompanied by morphological alterations. Shikonin effectively reversed these adverse effects by inhibiting PKM2 tetramerization, rescuing the loss of osteogenic function in BMSCs. The therapeutic potential of shikonin was confirmed in the diabetic periodontitis mouse model. CONCLUSION PKM2 impairs the osteogenesis of BMSCs by affecting metabolism and mitochondrial function, suggesting it as a potential therapeutic target for diabetic periodontitis.
Collapse
Affiliation(s)
- Yanlin Zhu
- College of Stomatology, Chongqing Medical University, 426# Songshibei Road, Yubei District, Chongqing, 401147, P.R. China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Edcation, Chongqing, China
| | - Yuhan Yang
- College of Stomatology, Chongqing Medical University, 426# Songshibei Road, Yubei District, Chongqing, 401147, P.R. China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Edcation, Chongqing, China
| | - Yuyan Lan
- College of Stomatology, Chongqing Medical University, 426# Songshibei Road, Yubei District, Chongqing, 401147, P.R. China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Edcation, Chongqing, China
| | - Zun Yang
- College of Stomatology, Chongqing Medical University, 426# Songshibei Road, Yubei District, Chongqing, 401147, P.R. China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Edcation, Chongqing, China
| | - Xiang Gao
- College of Stomatology, Chongqing Medical University, 426# Songshibei Road, Yubei District, Chongqing, 401147, P.R. China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Edcation, Chongqing, China
| | - Jie Zhou
- College of Stomatology, Chongqing Medical University, 426# Songshibei Road, Yubei District, Chongqing, 401147, P.R. China.
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Edcation, Chongqing, China.
| |
Collapse
|
27
|
Dovhalyuk V, Yang F, Nikolic S, Vujasinovic M, Löhr JM, Globisch D. Differences in the Fecal Metabolome of Autoimmune Pancreatitis Patients. United European Gastroenterol J 2025. [PMID: 40243134 DOI: 10.1002/ueg2.70025] [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: 09/08/2024] [Revised: 02/08/2025] [Accepted: 02/16/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Chronic pancreatitis is a risk factor for pancreatic cancer. Autoimmune pancreatitis is a unique form of chronic pancreatitis that is primarily characterized by its immune mediate etiology, clinically resembling pancreatic cancer, yet uniquely responsive to steroid treatment. OBJECTIVE Early and accurate diagnosis of autoimmune pancreatitis is vital for effective treatment and patient prognosis, for which new diagnostic tools are urgently required. Gut microbiota dysbiosis has been identified to correlate with the development of pancreatic diseases, which provides new opportunities for the discovery of disease biomarkers. METHODS We utilized a mass spectrometric global metabolomics investigation of patient autoimmune pancreatitis and chronic pancreatitis fecal samples, investigating microbiome, dietary and human metabolism. RESULTS We discovered a series of newly identified metabolic signatures between both patient groups including enterolactone, 4-guanidinobutanoic acid, and methylthioadenosine sulfoxide. Additionally, the analysis revealed significant differences in several metabolic pathways such as fatty acids, alkaloids, amino acids and peptides. CONCLUSION Our observations provide novel insights into important metabolic human pathways and microbiome-derived metabolites to distinguish autoimmune pancreatitis from chronic pancreatitis. These findings reveal systemic metabolic responses and the identified metabolites may be developed into potential biomarkers for future diagnosis to distinguish between autoimmune pancreatitis and chronic pancreatitis.
Collapse
Affiliation(s)
- Vladyslav Dovhalyuk
- Department of Chemistry - BMC, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Fan Yang
- Department of Chemistry - BMC, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Sara Nikolic
- Department of Gastroenterology, Clinic of Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia
- Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Miroslav Vujasinovic
- Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
- Department for Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - J-Matthias Löhr
- Department for Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - Daniel Globisch
- Department of Chemistry - BMC, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
28
|
Zhu S, Li J, Sun H, Liang J, Qiu Z, Zhou X, Wang W, Wei D, Zhong L. A biotin guided Pt IV amphiphilic prodrug synergized with CDK4/6 inhibition for enhanced tumor targeted therapy. NANOSCALE 2025; 17:9907-9913. [PMID: 40136056 DOI: 10.1039/d5nr00218d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Platinum-based chemotherapy has been the first-line treatment for advanced bladder cancer for decades, but its durability and safety remain important challenges. Targeted delivery and other precision medicine bring hope to fight cancer. In this study, we present a novel targeted therapy utilizing a biotin receptor-targeting lipid PtIV prodrug amphiphile, which encapsulates a CDK4/6 inhibitor into BPtIV@Rib. CDK4/6 inhibitors have the potential to combat breast cancer and enhance sensitivity to cisplatin, thereby improving its therapeutic efficacy. Our findings demonstrate that BPtIV@Rib also exhibits excellent bladder tumor-targeting capability, resulting in increased accumulation of Pt and ribociclib (Rib) at the tumor site. The combination of PtIV and Rib leads to substantial tumor growth suppression while minimizing synergistic toxicity compared to conventional therapies. In conclusion, this combination therapy represents a promising strategy for enhanced targeted treatment of bladder cancer, potentially improving patient outcomes while reducing adverse effects.
Collapse
Affiliation(s)
- Shaoming Zhu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Jiaxu Li
- College of Chemistry and Materials, Graduate School, Nanning Normal University, Nanning 530001, People's Republic of China
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Hao Sun
- Department of Breast Surgery, Sixth Affiliated Hospital of Harbin Medical University, Harbin 150023, China.
| | - Jian Liang
- Department of Breast Surgery, Sixth Affiliated Hospital of Harbin Medical University, Harbin 150023, China.
| | - Zhi Qiu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Xiaoguang Zhou
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Wei Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Dengshuai Wei
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Lei Zhong
- Department of Breast Surgery, Sixth Affiliated Hospital of Harbin Medical University, Harbin 150023, China.
| |
Collapse
|
29
|
Liu L, Xing G, Guo X, Chen H, Li J, Wang J, Li Y, Liang G, Liu M. Inhibition of colorectal cancer cell growth by downregulation of M2-PK and reduction of aerobic glycolysis by clove active ingredients. Front Pharmacol 2025; 16:1552486. [PMID: 40308769 PMCID: PMC12041220 DOI: 10.3389/fphar.2025.1552486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 03/24/2025] [Indexed: 05/02/2025] Open
Abstract
Exploring the anti-tumor molecular mechanisms of traditional Chinese medicines has become an important strategy to develop novel anti-tumor drugs in the clinic. Several pharmacological studies have reported the antioxidant, antibacterial, anti-inflammatory, and anti-tumor effects of clove. Previously, we have shown that the active fraction from clove (AFC) can inhibit the growth of tumor cells, particularly colon cancer cells, in vitro. However, the mechanism of action regarding the anti-colon cancer activity of AFC, especially in aerobic glycolysis, has not been adequately investigated. In this study, we found that AFC significantly inhibited the growth of five types of colon cancer cells, downregulated the mRNA and protein levels of M2-type pyruvate kinase (PKM2), and reduced aerobic glycolysis capacity. Transfection of PKM2-siRNA mimicked the inhibitory effects of AFC on aerobic glycolysis in colon cancer cells. Furthermore, the highly expressed, tumor-specific targets c-myc and cyclin D1 in cells were also found to be downregulated following the action of AFC. In the HCT116 cell xenograft nude mice models, the results after AFC administration were consistent with those of the cellular experiments, while AFC caused less liver injury and weight loss than the conventional chemotherapeutic agent 5- fluorouracil (5-FU). In conclusion, AFC inhibits colon cancer growth by downregulating PKM2 to inhibit aerobic glycolysis and reduce the tumor-specific high expression of c-myc and cyclin D1. Future work should explore how it downregulates pyruvate kinase (PK) in the first place, along with the intrinsic mechanism between the downregulation of PKM2 and the downregulation of c-myc.
Collapse
Affiliation(s)
- Lin Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Drug Dispending Department, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, China
| | - Gang Xing
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaoyi Guo
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jian Li
- Pharmacy Department, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Jian Wang
- Discipline Construction Office, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yaling Li
- Pharmacy Department, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Gang Liang
- Pharmacy Department, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Minghua Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
30
|
Xi Y, Huang Y, Hu J, Wang Y, Qian Q, Tu L, Nie H, Zhu J, Ding C, Gao X, Zheng X, Huang D, Cheng L. EIF2B5 promotes malignant progression of hepatocellular carcinoma by activating the PI3K/AKT signaling pathway through targeting RPL6. Cell Signal 2025; 132:111821. [PMID: 40246131 DOI: 10.1016/j.cellsig.2025.111821] [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: 11/26/2024] [Revised: 04/05/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
Hepatocellular carcinoma (HCC) is a highly aggressive malignancy with limited treatment options and poor prognosis. In this study, we demonstrated the critical role of EIF2B5 in driving HCC progression. We found EIF2B5 expression is significantly upregulated in HCC tumor tissues in several bioinformatics datasets, including The Cancer Genome Atlas, and that high expression of EIF2B5 predicts poor prognosis for HCC patients. Through a series of in vitro cell biology experiments, we found that EIF2B5 knockdown significantly attenuated Hep3B and HepG2 proliferation, migration, and invasion and increased cell cycle arrest, whereas EIF2B5 overexpression promoted HCC progression. Through mass spectrometry and immunoprecipitation validation, we found that EIF2B5 directly interacted with RPL6 and that when EIF2B5 was overexpressed in HCC cells, it promoted the expression of the downstream protein RPL6, which was able to activate the phosphatidylinositol kinase (PI3K)/serine-threonine kinase (AKT)/mammalian target of rapamycin (mTOR) pathway and thereby increase the proliferation and invasion ability of HCC cell lines, as verified by second-generation sequencing analysis and western blot. We further verified these findings using the mouse ectopic tumor assay, and the results showed that EIF2B5 knockdown significantly inhibited tumor progression in HCC mice. The present study suggests that EIF2B5 promotes malignant progression of HCC by interacting with RPL6 and activating the PI3K/AKT/mTOR signaling pathway and may serve as a potential target for the treatment of HCC.
Collapse
Affiliation(s)
- Yiling Xi
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yue Huang
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiahui Hu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yan Wang
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qiyi Qian
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Linglan Tu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Huizong Nie
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiayao Zhu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chenguang Ding
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaotao Gao
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaoliang Zheng
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Dongsheng Huang
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Liyan Cheng
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
31
|
Wang B, Jiang C, Yu P, Nie Z, Wang N, Zhang X. Curvilinear relationship between life's crucial 9 and metabolic syndrome in U.S. adults: a cross-sectional study. Front Endocrinol (Lausanne) 2025; 16:1559413. [PMID: 40303636 PMCID: PMC12039309 DOI: 10.3389/fendo.2025.1559413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Background Metabolic Syndrome (MetS) is closely linked to cardiovascular disease. However, no studies have examined the relationship between Life's Crucial 9 (LC9) and MetS. Our goal is to investigate the potential association between LC9 and MetS. Methods We employed a weighted multivariate logistic regression model to evaluate the relationship between LC9, health behavior score, health factors score, and MetS. To assess the robustness of this association, we conducted sensitivity analyses. Furthermore, we utilized smooth curve fitting to investigate the potential curvilinear relationships between LC9, health behavior score, health factors score, and MetS. To pinpoint inflection points, we integrated recursive partitioning algorithms with a two-stage linear regression model. Additionally, we performed stratified analyses to explore heterogeneity across different population subgroups. Results Our study included a total of 28,555 participants. In the regression model that accounted for all covariates, the OR for LC9 and MetS was 0.941 (0.939, 0.944), indicating a significant negative correlation between the two. Smooth curve analysis confirmed a curvilinear relationship between LC9 and MetS, with an inflection point at 70.56. The negative correlation was evident both before and after the inflection point, with a more pronounced effect after the inflection point. Subgroup analyses of Health behavior score and Health factors score, as well as stratified analyses by age, sex, and BMI, showed that all groups exhibited curvilinear relationships consistent with the overall pattern. Conclusion The curvilinear relationship between LC9 scores and metabolic syndrome indicates that higher LC9 scores act as a protective factor against MetS.
Collapse
Affiliation(s)
- Bo Wang
- Central Hospital of Jinan City, Jinan, Shandong, China
| | - Chunqi Jiang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Pingping Yu
- Wendeng District People’s Hospital of Weihai City, Wendeng District, weihai, Shandong, China
| | - Zhen Nie
- Wendeng District People’s Hospital of Weihai City, Wendeng District, weihai, Shandong, China
| | - Ning Wang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xin Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
32
|
Tsunematsu M, Shirai Y, Hamura R, Haruki K, Furukawa K, Onda S, Matsumoto M, Okui N, Taniai T, Ikegami T. The conventional method of blood-loss calculation can underestimate true blood loss during laparoscopic pancreaticoduodenectomy: a dual-institute experience. Surg Today 2025:10.1007/s00595-025-03040-y. [PMID: 40232398 DOI: 10.1007/s00595-025-03040-y] [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: 11/15/2024] [Accepted: 03/03/2025] [Indexed: 04/16/2025]
Abstract
PURPOSE This study investigated whether blood loss (BL) was. truly less in patients undergoing laparoscopic pancreaticoduodenectomy (LPD) than in those undergoing open pancreaticoduodenectomy (OPD). METHODS A total of 98 patients undergoing pancreaticoduodenectomy between 2021 and 2023 were divided into OPD and LPD groups and compared. Estimated BL was calculated from the perioperative changes in hematocrit, hemoglobin, or red blood cell volume. Actual BL was calculated using the conventional method as the sum of the intraoperative aspirated blood volume and surgical gauze weight. RESULTS Actual BL was significantly lower in the LPD group than in the OPD group (150 [80-350] ml vs. 345 [150-700] ml, p = 0.003). However, there were no significant differences in the estimated BL calculated from the hematocrit (461 [187-626] ml vs. 351 [153-737] ml, p = 0.972), hemoglobin, or red blood cell volume. Estimated BL showed a stronger linear correlation with actual BL in the OPD group (r = 0.447-0.669) than in the LPD group (r = 0.158-0.417). OPD was not a significant factor in the increased estimated BL. CONCLUSIONS The conventional method for calculating the BL in LPD may underestimate the actual loss, highlighting the need for a more accurate method of evaluation.
Collapse
Affiliation(s)
- Masashi Tsunematsu
- Department of Digestive Surgery, Saku Central Hospital Advanced Care Center, 3400-28, Nakagomi, Saku, Nagano, 385-0051, Japan.
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan.
| | - Yoshihiro Shirai
- Department of Digestive Surgery, Saku Central Hospital Advanced Care Center, 3400-28, Nakagomi, Saku, Nagano, 385-0051, Japan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Ryoga Hamura
- Department of Digestive Surgery, Saku Central Hospital Advanced Care Center, 3400-28, Nakagomi, Saku, Nagano, 385-0051, Japan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Koichiro Haruki
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Kenei Furukawa
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Shinji Onda
- Department of Digestive Surgery, Saku Central Hospital Advanced Care Center, 3400-28, Nakagomi, Saku, Nagano, 385-0051, Japan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Michinori Matsumoto
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Norimitsu Okui
- Department of Digestive Surgery, Saku Central Hospital Advanced Care Center, 3400-28, Nakagomi, Saku, Nagano, 385-0051, Japan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Tomohiko Taniai
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Toru Ikegami
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| |
Collapse
|
33
|
Li YE, Liu S, Wang L, Du Y, Wu L, Chen H, Zhu T, Lin J, Xiong S, Wang Y, Zheng Q, Zou R, Lin L, Li Z, Wang L, Ge J, Ren J, Zhang Y. March2 Alleviates Aortic Aneurysm/Dissection by Regulating PKM2 Polymerization. Circ Res 2025; 136:e73-e93. [PMID: 40079144 DOI: 10.1161/circresaha.124.325049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 02/19/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Aortic aneurysm/dissection (AAD) is a life-threatening disease lacking effective pharmacological treatment. Protein ubiquitination plays a pivotal role in cardiovascular diseases. However, the possible contribution of the E3 ubiquitin ligase March2 (membrane-associated RING [really interesting new gene] finger protein 2) to the cause of AAD remains elusive. METHODS Integrated single-cell RNA sequencing analysis was conducted in human AAD tissues. Based on the screening results, we generated a mouse line of smooth muscle cell-specific March2 knockout. β-Aminopropionitrile monofumarate was used to establish AAD. Cleavage under targets and tagmentation and cleavage under targets and tagmentation-quantitative polymerase chain reaction were performed to identify possible target genes for histone H3K18 lactylation. RESULTS March2 expression was downregulated in aorta from patients with AAD or β-aminopropionitrile monofumarate-induced AAD mice. β-Aminopropionitrile monofumarate-induced AAD was significantly accentuated in March2 global (March2-/-) and vascular smooth muscle cell-specific deletion (March2fl/fl; TaglnCre) mice, whereas the AAD pathology was rescued by rAAV9-SM22α (smooth muscle 22α)-March2 (recombinant adeno-associated virus serotype 9 expressing Flag-tagged March2 under SM22α promoter). March2 interacted with PKM2 (pyruvate kinase M2) to promote K33-linked polyubiquitination. Deficiency of March2 lessened PKM2 dimer-to-tetramer conversion in AAD and overtly exacerbated AAD-induced histone H3K18 lactylation in vascular smooth muscle cells by fostering glucose metabolism reprogramming, thereby promoting p53-driven apoptotic transcriptional response-a hallmark of AAD pathogenesis. TEPP-46 (tetraethyl pyrophosphate), a PKM2-specific activator, pronouncedly alleviated March2 deficiency-deteriorated AAD pathology. CONCLUSIONS Our findings demonstrated that March2 is a novel endogenous defender that prevents AAD by inhibiting vascular smooth muscle cell apoptosis, suggesting that March2 represents a potential therapeutic target for AAD.
Collapse
Affiliation(s)
- Yiran E Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Shuolin Liu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- Department of Vascular Surgery (Z.L., Lixin Wang), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Litao Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Yuxin Du
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Lin Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Haoran Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Tingfang Zhu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Jie Lin
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Shengjun Xiong
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Yayu Wang
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Guangdong, China (Y.W., Q.Z.)
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, Guangdong (Y.W.)
| | - Qijun Zheng
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Guangdong, China (Y.W., Q.Z.)
| | - Rongjun Zou
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine (R.Z.)
| | - Ling Lin
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Zheyun Li
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- Department of Vascular Surgery (Z.L., Lixin Wang), Zhongshan Hospital, Fudan University, China
- Vascular Surgery Institute of Fudan University, Shanghai, China (Z.L., Lixin Wang)
| | - Lixin Wang
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- Department of Vascular Surgery (Z.L., Lixin Wang), Zhongshan Hospital, Fudan University, China
- Vascular Surgery Institute of Fudan University, Shanghai, China (Z.L., Lixin Wang)
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Yingmei Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| |
Collapse
|
34
|
Zhao F, Zhu L, Lu Y, Xia H, Zhang S, Zhang P. A prospective study of regular milk drinking and the risk of all-cause and cause-specific mortality in U.S. adults: findings from the NHANES 2003-2008. NUTR HOSP 2025. [PMID: 40326324 DOI: 10.20960/nh.05623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND scholars have studied the influences of milk consumption on health. This prospective cohort study evaluated the association between regular milk consumption (RMD) and mortality, including all-cause, cardiovascular disease (CVD), and cancer-specific mortality, in Americans who were aged 20 years and older. METHODS it was attempted to analyze data from the 2003 to 2008 National Health and Nutrition Examination Survey (NHANES), including 8,653 participants with an average follow-up of 160.59 months (standard deviation = 0.96). Regular milk consumption (RMD) was defined as drinking milk at least five times per week. Cox proportional hazards regression models were thereafter utilized for analysis. RESULTS a significant association was noticeable particularly between RMD and higher rates of all-cause mortality (hazard ratio (HR) = 1.27; 95 % confidence interval (CI): 1.03-1.56; p = 0.03) and cancer mortality (HR = 1.53; 95 % CI: 1.01-2.32; p = 0.04), rather than with CVD mortality (HR = 1.03; 95 % CI: 0.73-1.44; p = 0.84). The association between RMD and all-cause mortality was notably observed across various subgroups in sensitivity and subgroup analyses. CONCLUSIONS these outcomes unveiled the importance of exercising caution with milk consumption, which could be emphasized in public health recommendations.
Collapse
Affiliation(s)
- Fei Zhao
- Department of Nursing. Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University
| | - Liyan Zhu
- Department of Nursing. Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University
| | - Ye Lu
- Department of Nursing. Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University
| | - Hongwei Xia
- Department of Thoracic Surgery. Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University
| | - Shengchao Zhang
- Department of Thoracic Surgery. Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University
| | - Ping Zhang
- Department of Medical Affairs. Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University
| |
Collapse
|
35
|
Sufiyan S, Salam H, Ilyas S, Amin W, Arshad F, Fatima K, Naeem S, Laghari AA, Enam SA, Mughal N. Prognostic implications of DNA methylation machinery (DNMTs and TETs) expression in gliomas: correlations with tumor grading and patient survival. J Neurooncol 2025:10.1007/s11060-025-05032-x. [PMID: 40208514 DOI: 10.1007/s11060-025-05032-x] [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: 01/28/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025]
Abstract
PURPOSE DNA methylation is a crucial epigenetic modification that regulates gene expression and chromatin structure. Its dysregulation is linked to glioma progression and prognosis, particularly through alterations in methylation machinery. DNMTs and TETs play key roles in these processes, but their involvement in gliomagenesis remains complex, especially in the context of IDH mutations. This study examines the expression patterns of DNMT and TET family genes in gliomas to assess their prognostic significance and therapeutic potential. MATERIALS AND METHODS mRNA expression levels of DNMT1, DNMT3A, DNMT3B, DNMT3L, TET1, TET2, TET3, and TDG were analyzed in 75 glioma samples and 10 normal controls using real-time quantitative PCR (qPCR). Statistical analyses and graphical representation were performed using R (v3.3.2) and RStudio (v1.4.1717), with p-values < 0.05 considered significant. Findings were validated using publicly available databases, TCGA and CGGA. RESULTS Compared to normal controls, DNMTs and TETs were significantly downregulated in gliomas, with expression levels inversely correlated with histological grade. Survival analysis using the log-rank test demonstrated a significant association between lower TETs and DNMTs expression and an increased risk of mortality. However, multivariate Cox regression analysis indicated that DNMTs and TETs expression were not independent prognostic markers for patient survival, suggesting their impact may be influenced by other clinical and molecular factors. Validation through online databases (TCGA and CGGA) showed that TET family expression across histological grades was consistent with our samples, whereas TDG and DNMT family expression differed. CONCLUSION Our findings suggest that DNMTs and TETs may serve as therapeutic targets in glioma due to their downregulation and association with survival, with TET family members (TET1, TET2, and TET3) validated through online databases. However, their prognostic value is limited, as other clinical and molecular factors influence patient outcomes. The downregulation of DNMTs in our samples compared to online databases can be attributed to distinct epigenetic mechanisms: in IDH-mutant gliomas, DNMT suppression results from global hypermethylation (G-CIMP) due to 2-HG accumulation, which inhibits TET enzymes and disrupts DNA methylation homeostasis. In contrast, IDH-wildtype high-grade gliomas exhibit global hypomethylation, genomic instability, oncogenic signaling, and dedifferentiation, reducing the demand for active DNA methylation maintenance. These findings underscore the complex regulation of DNMTs and TETs in gliomas and their potential therapeutic implications.
Collapse
Affiliation(s)
- Sufiyan Sufiyan
- Department of Surgery, Aga Khan University Hospital, Karachi, Pakistan
| | - Hira Salam
- Department of Oral Pathology, Dr. Ishrat-ul-Ibad Khan Institute of Oral Health Sciences, Dow University of Health Sciences, Karachi, Pakistan
| | - Sahar Ilyas
- Center of Oncological Research in Surgery, Aga Khan University, Karachi, Pakistan
| | - Wajiha Amin
- Department of Surgery, Aga Khan University Hospital, Karachi, Pakistan
| | - Fatima Arshad
- Department of Pathology, Dow International Medical College, Dow University of Health Sciences, Karachi, 75300, Pakistan
| | | | - Sana Naeem
- Center of Oncological Research in Surgery, Aga Khan University, Karachi, Pakistan
| | - Altaf Ali Laghari
- Department of Surgery, Aga Khan University Hospital, Karachi, Pakistan
| | - Syed Ather Enam
- Department of Surgery, Aga Khan University Hospital, Karachi, Pakistan.
- Center of Oncological Research in Surgery, Aga Khan University, Karachi, Pakistan.
- Centre for Regenerative Medicine and Stem Cell Research, Aga Khan University, Karachi, Pakistan.
| | - Nouman Mughal
- Department of Biological & Biomedical Science, Aga Khan University Hospital, Karachi, Pakistan.
- Center of Oncological Research in Surgery, Aga Khan University, Karachi, Pakistan.
| |
Collapse
|
36
|
Xiong Y, Du Y, Lin F, Fu B, Guo D, Sha Z, Tian R, Yao R, Wang L, Cong Z, Li B, Lin X, Wu H. SENP1-SIRT3 axis mediates glycolytic reprogramming to suppress inflammation during Listeria monocytogenes infection. mBio 2025; 16:e0252424. [PMID: 40071948 PMCID: PMC11980586 DOI: 10.1128/mbio.02524-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 02/12/2025] [Indexed: 04/10/2025] Open
Abstract
Listeria monocytogenes, a foodborne pathogen, has the ability to invade intestinal mucosal cells, undergo intracellular proliferation, activate host immune responses, and induce diseases such as colitis. We have demonstrated that sentrin-specific protease 1 (SENP1) functions as a protective gene in the host, suppressing the inflammatory response triggered by Listeria monocytogenes. The host's SENP1-SIRT3 axis plays a critical role in regulating inflammation during Listeria monocytogenes infection. Our findings reveal that overexpression of SENP1, particularly under Listeria monocytogenes infection conditions (MOI = 20), effectively suppresses inflammation through modulation of glycolysis. Mechanistically, during Listeria monocytogenes infection, SENP1 accumulates in the mitochondria, facilitating the de-SUMOylation and activation of sirtuin 3 (SIRT3). Activated SIRT3 then regulates the deacetylation of pyruvate kinase M2 (PKM2), leading to a decrease in glycolytic intermediates, downregulation of glycolysis-related gene expression, and suppression of inflammation. Taken together, our study provides a deeper understanding of the mechanistic role of the SENP1-SIRT3 axis in the regulation of inflammation, offering novel insights, and strategies for the treatment and prevention of inflammatory diseases. IMPORTANCE Sentrin-specific protease 1 (SENP1)-sirtuin 3 (SIRT3) has never been reported in the regulation of bacteria-induced inflammation. Our study demonstrated that SENP1 acted as a protective factor against Listeria-induced inflammation by promoting SIRT3 activation and subsequent metabolic reprogramming. The SENP1-SIRT3 axis served not only as an essential signaling pathway for regulating mitochondrial metabolic responses to metabolic stress but also responds to bacterial invasion and plays a protective role in the organism. Our findings provide a basis for further research into targeting the SENP1-SIRT3 signaling pathway for the treatment of bacterial infections.
Collapse
Affiliation(s)
- Yan Xiong
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Yongliang Du
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Feng Lin
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Beibei Fu
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Dong Guo
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Zhou Sha
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Rong Tian
- Department of Pathology, Chongqing Hygeia Hospital, Chongqing, China
| | - Rui Yao
- Department of Pathology, Chongqing Hygeia Hospital, Chongqing, China
| | - Lulu Wang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Zixuan Cong
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Bohao Li
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Xiaoyuan Lin
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Haibo Wu
- School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
37
|
Delobelle Q, Inizan TJ, Adjoua O, Lagardère L, Célerse F, Maréchal V, Piquemal J. High-Resolution Molecular-Dynamics Simulations of the Pyruvate Kinase Muscle Isoform 1 and 2 (PKM1/2). Chemistry 2025; 31:e202402534. [PMID: 39614705 PMCID: PMC11973853 DOI: 10.1002/chem.202402534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/01/2024]
Abstract
Glucose metabolism plays a pivotal role in physiological processes and cancer growth. The final stage of glycolysis, converting phosphoenolpyruvate (PEP) into pyruvate, is catalyzed by the pyruvate kinase (PK) enzyme. Whereas PKM1 is mainly expressed in cells with high energy requirements, PKM2 is preferentially expressed in proliferating cells, including tumor cells. Structural analysis of PKM1 and PKM2 is essential to design new molecules with antitumoral activity. To understand their structural dynamics, we performed extensive high-resolution molecular dynamics (MD) simulations using adaptive sampling techniques coupled to the polarizable AMOEBA force field. Performing more than 6 μs of simulation, we considered all oligomerization states of PKM2 and propose structural insights for PKM1 to further study the PKM2-specific allostery. We focused on key sites including the active site and the natural substrate Fructose Bi-Phosphate (FBP) fixation pocket. Additionally, we present the first MD simulation of biologically active PKM1 and uncover important similarities with its PKM2 counterpart bound to FBP. We also analysed TEPP-46's fixation, a pharmacological activator binding a different pocket, on PKM2 and highlighted the structural differences and similarities compared to PKM2 bound to FBP. Finally, we determined potential new cryptic pockets specific to PKM2 for drug targeting.
Collapse
Affiliation(s)
- Quentin Delobelle
- Centre de Recherche Saint-Antoine – Team “Biologie et Thérapeutique du Cancer”, UMRS 938 INSERMParisFrance
- Sorbonne Université, CNRS, Laboratoire de Chimie Théorique, UMR 761675005ParisFrance
| | - Théo Jaffrelot Inizan
- Sorbonne Université, CNRS, Laboratoire de Chimie Théorique, UMR 761675005ParisFrance
- University of California BerkeleyBakar Institute of Digital Materials for the PlanetCollege of Computing, Data Science, and SocietyBerkeley94720USA
| | - Olivier Adjoua
- Sorbonne Université, CNRS, Laboratoire de Chimie Théorique, UMR 761675005ParisFrance
| | - Louis Lagardère
- Sorbonne Université, CNRS, Laboratoire de Chimie Théorique, UMR 761675005ParisFrance
| | - Frédéric Célerse
- Sorbonne Université, CNRS, Laboratoire de Chimie Théorique, UMR 761675005ParisFrance
- Sorbonne Université, CNRS, IPCM75005ParisFrance
| | - Vincent Maréchal
- Centre de Recherche Saint-Antoine – Team “Biologie et Thérapeutique du Cancer”, UMRS 938 INSERMParisFrance
| | - Jean‐Philip Piquemal
- Sorbonne Université, CNRS, Laboratoire de Chimie Théorique, UMR 761675005ParisFrance
| |
Collapse
|
38
|
Li Y, Zong K, Zhou Y, Sun Y, Liu Y, Zhou B, Wu Z. Enhanced preoperative prediction of pancreatic fistula using radiomics and clinical features with SHAP visualization. Front Bioeng Biotechnol 2025; 13:1510642. [PMID: 40256777 PMCID: PMC12006764 DOI: 10.3389/fbioe.2025.1510642] [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: 10/13/2024] [Accepted: 03/21/2025] [Indexed: 04/22/2025] Open
Abstract
Background Clinically relevant postoperative pancreatic fistula (CR-POPF) represents a significant complication after pancreaticoduodenectomy (PD). Therefore, the early prediction of CR-POPF is of paramount importance. Based on above, this study sought to develop a CR-POPF prediction model that amalgamates radiomics and clinical features to predict CR-POPF, utilizing Shapley Additive explanations (SHAP) for visualization. Methods Extensive radiomics features were extracted from preoperative enhanced Computed Tomography (CT) images of patients scheduled for PD. Subsequently, feature selection was performed using Least Absolute Shrinkage and Selection Operator (Lasso) regression and random forest (RF) algorithm to select pertinent radiomics and clinical features. Last, 15 CR-POPF prediction models were developed using five distinct machine learning (ML) predictors, based on selected radiomics features, selected clinical features, and a combination of both. Model performance was compared using DeLong's test for the area under the receiver operating characteristic curve (AUC) differences. Results The CR-POPF prediction model based on the XGBoost predictor with the combination of the radiomics and clinical features selected by Lasso regression and RF exhibited superior performance among these 15 CR-POPF prediction models, achieving an accuracy of 0.85, an AUC of 0.93. DeLong's test showed statistically significant differences (P < 0.05) when compared to the radiomics-only and clinical-only models, with recall of 0.63, precision of 0.65, and F1 score of 0.64. Conclusion The proposed CR-POPF prediction model based on the XGBoost predictor with the combination of the radiomics and clinical features selected by Lasso regression and RF can effectively predicting the CR-POPF and may provide strong support for early clinical management of CR-POPF.
Collapse
Affiliation(s)
- Yan Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kenzhen Zong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yin Zhou
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuan Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yanyao Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Baoyong Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Hepatobiliary Surgery, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Zhongjun Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
39
|
de Alwis N, Beard S, Baird L, Binder NK, Pritchard N, Tong S, Kaitu'u-Lino TJ, Hui L, Hannan NJ. Phosphoglucomutase 5 gene transcripts are expressed by the human placenta and differentially regulated in placental dysfunction. Sci Rep 2025; 15:11381. [PMID: 40180976 PMCID: PMC11968825 DOI: 10.1038/s41598-025-94498-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
The placenta plays an essential role facilitating nutrient, gas and waste exchange between the maternal and fetal systems for optimal fetal growth. When placental development is impaired and the placenta dysfunctional, serious pregnancy complications such as fetal growth restriction and preeclampsia may arise. Previously, phosphoglucomutase-5 (PGM5) transcripts were found to be highly elevated in the blood of patients whose pregnancies were complicated by fetal growth restriction and preeclampsia. As both conditions feature placental insufficiency, here we aimed to characterise PGM5 levels in the healthy and dysfunctional placenta. PGM5 expression was detectable in all placental samples across gestation, in cases of preterm preeclampsia, fetal growth restriction and controls. PGM5 mRNA expression was significantly downregulated in the pathological placentas compared to controls, but PGM5 protein production was not dysregulated. Isolated cytotrophoblast and placental explant tissue exposed to hypoxia (modelling placental dysfunction) demonstrated significantly increased PGM5 expression, but again did not change protein levels. Silencing PGM5 expression under hypoxic conditions in primary cytotrophoblast did not alter anti-angiogenic sFLT-1 secretion but increased expression of multiple genes associated with cell growth, apoptosis and oxidative stress, whilst also increasing cell viability. Expression of PGM5 in all placental samples assessed suggests that PGM5 has functions in the placenta. However, further investigation could be performed to explore the discrepancies in protein and mRNA expression, as well as the precise function of PGM5 in the placenta, and whether altered PGM5 levels may be important for placental development.
Collapse
Affiliation(s)
- Natasha de Alwis
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
- Northern Health, Epping, Melbourne, VIC, 3076, Australia
| | - Sally Beard
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
- Northern Health, Epping, Melbourne, VIC, 3076, Australia
| | - Lydia Baird
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
- Northern Health, Epping, Melbourne, VIC, 3076, Australia
| | - Natalie K Binder
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
| | - Natasha Pritchard
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
| | - Stephen Tong
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
| | - Lisa Hui
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
- Northern Health, Epping, Melbourne, VIC, 3076, Australia
| | - Natalie J Hannan
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia.
- Northern Health, Epping, Melbourne, VIC, 3076, Australia.
| |
Collapse
|
40
|
Luo YY, Guan YP, Zhan HF, Sun CY, Cai LY, Tao KG, Lin Y, Zeng X. Circ_0098181 binds PKM2 to attenuate liver fibrosis. Front Pharmacol 2025; 16:1517250. [PMID: 40248098 PMCID: PMC12003362 DOI: 10.3389/fphar.2025.1517250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/14/2025] [Indexed: 04/19/2025] Open
Abstract
Background Liver cirrhosis seriously harms human health and fibrosis is the essential pathological process of cirrhosis. Recently, circular RNAs (circRNAs) were found to play critical roles in liver fibrosis, but the key circRNAs and precise mechanisms remained unclear. This study aimed to investigate the effect of circ_0098181 in fibrogenesis and explore its mechanism. Methods RNA sequencing was conducted to identify circRNA signatures in human liver cirrhotic tissues. Hepatic stellate cells (HSCs) (including primary rat HSCs, LX2, HSC-T6) and carbon tetrachloride (CCl4) induced liver cirrhosis model were used to explore the role of circ_0098181 on HSC activation and liver fibrogenesis in vitro and in vivo. RNA sequencing, RNA pull-down, mass spectrometry, and RNA immunoprecipitation (RIP) experiments were performed to elucidate the mechanism. Results Circ_0098181 was obviously reduced in human fibrotic liver tissues and activated HSCs. Exogenous administration of circ_0098181 blocked the activation, proliferation, and migration of HSCs in vitro and mitigated the progression of CCl4-induced liver fibrosis in vivo. Mechanistically, adenosine deaminase acting on RNA1 (ADAR1) combined with the intronic complementary sequences (ICSs) in the flanking regions, thereby regulating the biogenesis of circ_0098181. RNA sequencing and qRT-PCR revealed the suppression of circ_0098181 on pro-inflammation cytokines expression (TNFα, Fas, Cxcl11, etc.). RNA pull-down, mass spectrometry, and RIP experiments indicated that pyruvate kinase M2 (PKM2) was the direct target of circ_0098181. Circ_0098181 bound to PKM2, restrained its nuclear translocation and phosphorylation. Conclusion In conclusion, circ_0098181 exerts a significant anti-fibrotic effect by binding PKM2 to repress its nuclear translocation and inhibiting hepatic inflammation, suggesting the promising therapeutic merit in liver cirrhosis.
Collapse
Affiliation(s)
- Yuan-Yuan Luo
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ya-Ping Guan
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hong-Fei Zhan
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chun-Yan Sun
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ling-Yan Cai
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ke-Gong Tao
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yong Lin
- Department of Gastroenterology, Shanghai Changzheng Hospital, Navy Military Medical University, Shanghai, China
| | - Xin Zeng
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
41
|
Song H, Gao M, Bao Z, Yang Y, Chen Y, Hao J. Expression and clinical significance of ITGA3 in breast cancer. Open Med (Wars) 2025; 20:20241113. [PMID: 40181838 PMCID: PMC11967466 DOI: 10.1515/med-2024-1113] [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: 09/09/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 04/05/2025] Open
Abstract
Background Integrin subunit Alpha 3 is essential for cell adhesion and movement, but its role in breast cancer (BC) is unclear. This study evaluated ITGA3 expression in BC and its clinical significance. Methods The Human Protein Atlas (HPA), UALCAN, and Kaplan-Meier plotter were used to analyze ITGA3 in BC. ITGA3 was evaluated using receiver operating characteristic curves. The cell counting kit-8 test was used to examine the role of ITGA3 in cell proliferation. Results BC tissues' ITGA3 protein and mRNA levels were significantly lower than normal controls. ITGA3 was associated with better recurrence-free survival (RFS) and distant metastasis-free survival, especially in estrogen receptor (ER)-positive, Luminal B, and Luminal B subtypes. ITGA3 predicted RFS for 5 years and the response to chemotherapy. ITGA3 was associated with ER status but not age, tumor, node, metastasis stages, or tumor size. ITGA3 has a positive impact on BC cell growth. Conclusions ITGA3 may be a predictive marker for BC and a therapeutic target. These findings need to be confirmed, and the molecular mechanisms of ITGA3 need to be clarified.
Collapse
Affiliation(s)
- Huachun Song
- Department of Thyroid Breast Oncology, Yiwu Central Hospital, Yiwu, 322099, Zhejiang, China
| | - Meihui Gao
- Clinical Oncology College, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Zheng Bao
- Operating Room, Yiwu Central Hospital, Yiwu, 322099, Zhejiang, China
| | - Yong Yang
- Department of Pathology, Yiwu Central Hospital, Yiwu, 322099, Zhejiang, China
| | - Yang Chen
- Department of Thyroid Breast Oncology, Yiwu Central Hospital, Yiwu, 322099, Zhejiang, China
| | - Jing Hao
- Department of Thyroid Breast Oncology, Yiwu Central Hospital, No. 519, Nanmen Street, Yiwu, 322099, Zhejiang, China
| |
Collapse
|
42
|
Mortazavi Farsani SS, Soni J, Jin L, Yadav AK, Bansal S, Mi T, Hilakivi-Clarke L, Clarke R, Youngblood B, Cheema A, Verma V. Pyruvate kinase M2 activation reprograms mitochondria in CD8 T cells, enhancing effector functions and efficacy of anti-PD1 therapy. Cell Metab 2025:S1550-4131(25)00106-8. [PMID: 40199327 DOI: 10.1016/j.cmet.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/27/2024] [Accepted: 03/06/2025] [Indexed: 04/10/2025]
Abstract
Mitochondria regulate T cell functions and response to immunotherapy. We show that pyruvate kinase M2 (PKM2) activation enhances mitochondria-dependent effector functions in CD8 and chimeric antigen receptor (CAR)-T cells. Multi-omics and 13C-glucose tracer studies showed that PKM2 agonism alters one-carbon metabolism, decreasing methionine levels, resulting in hypomethylated nuclear and mitochondrial DNA and enhancing mitochondrial biogenesis and functions. PKM2 activation increased the recall responses and anti-tumor functions of CD8 T cells, enhancing adoptive cell therapy. In preclinical models, the PKM2 agonist induced CD8 T cell-dependent anti-tumor responses that synergized with anti-programmed death 1 (PD1) therapy. Immunologically, PKM2 agonists boosted the activation of effector T cells while reducing FoxP3+ T regulatory (Treg) cells in the tumors. The anti-PD1 combination enhanced the frequency of tumor-specific activated CD8 T cells. Together, PKM2 agonism increased mitochondrial functions supporting cell cytotoxicity. Hence, pharmacological targeting of PKM2 can be a clinically viable strategy for enhancement of adoptive cell therapy, in situ anti-tumor immune responses, and immune checkpoint blockade therapy. VIDEO ABSTRACT.
Collapse
Affiliation(s)
| | - Jignesh Soni
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Lu Jin
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Anil Kumar Yadav
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Shivani Bansal
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Tian Mi
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | - Robert Clarke
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Benjamin Youngblood
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Amrita Cheema
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Vivek Verma
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
43
|
Yang J. Unveiling the multifaceted roles of long non-coding RNA CTBP1-DT in human diseases: Special attention to its microprotein-encoding potential. Pathol Res Pract 2025; 268:155870. [PMID: 40020329 DOI: 10.1016/j.prp.2025.155870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/30/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
C-terminal binding protein 1 divergent transcript (CTBP1-DT) is a novel long non-coding RNA (lncRNA) located on human chromosome 4p16.3. Numerous studies have shown that CTBP1-DT plays a critical regulatory role in various human malignancies and non-malignant diseases. In several cancers, the expression of CTBP1-DT is upregulated, closely associated with the risk of 12 types of cancer, and strongly correlated with the clinical pathological features and poor prognosis of 10 of these cancers. Mechanistically, CTBP1-DT is stimulated by the transcription factors ETV5 and Sp1, or methylated by YTHDC1. By competitively inhibiting 12 microRNAs, it activates 3 signaling pathways that influence malignant behaviors of tumor cells, including proliferation, apoptosis, cell cycle arrest, migration, invasion, immune evasion, and chemoresistance. Importantly, it also encodes the microprotein DNA damage up-regulated protein (DDUP), which mediates cisplatin resistance through sustained response to DNA damage signals. Furthermore, CTBP1-DT has been implicated in the progression of non-malignant diseases such as diabetes and related conditions, cardiovascular diseases, and osteoarthritis. This review summarizes the latest research on the RNA and protein functions of CTBP1-DT in human diseases, outlines various molecular regulatory networks centered around CTBP1-DT, and discusses the opportunities and challenges of its clinical applications.
Collapse
Affiliation(s)
- Jingjie Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin 300060, China.
| |
Collapse
|
44
|
Saegusa Y, Imaoka Y, Ohira M, Kobayashi T, Honmyo N, Hamaoka M, Onoe T, Takei D, Oishi K, Abe T, Nakayama T, Akabane M, Sasaki K, Ohdan H. Cluster analysis of hepatocellular carcinoma prognosis using preoperative alpha-fetoprotein and des-gamma-carboxy prothrombin levels: a multi-institutional study. J Gastrointest Surg 2025; 29:101980. [PMID: 39884550 DOI: 10.1016/j.gassur.2025.101980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) remains the leading cause of cancer-related mortality worldwide and is characterized by high recurrence rates after curative resection. The tumor markers des-gamma-carboxy prothrombin (DCP) and alpha-fetoprotein (AFP) are crucial for HCC diagnosis and prognosis. However, their roles in the modern era of HCC epidemiology require reevaluation. METHODS This multi-institutional retrospective study analyzed 1515 patients who underwent hepatectomy for primary HCC. Patients were classified into 4 clusters using k-means analysis based on preoperative DCP and AFP levels. Clinicopathologic characteristics, overall survival (OS), and recurrence rate (RR) were evaluated using Cox proportional hazards models and area under the receiver operating characteristic curve (AUROC) comparisons. RESULTS Cluster 3 (concurrent elevations of DCP and AFP) had the poorest 5-year OS (52.8%) and the highest RR (79.3%), whereas cluster 4 (low levels of both markers) had the most favorable outcomes, with a 5-year OS rate of 71.5% and an RR of 55.7%. Cluster 1 (elevated DCP alone) was associated with larger tumors (median of 45 mm) and more frequent vascular invasion (43%) than cluster 2 (elevated AFP alone, median tumor size of 24 mm, and vascular invasion of 36%). DCP was a stronger predictor of 5-year OS in patients with preserved liver function (AUROC, 0.63), whereas AFP was more effective in stratifying RR in patients with impaired liver function (AUROC, 0.57). Non-B, non-C hepatitis (NBNC)-related HCC exhibited a distinct biomarker profile, with an elevated DCP level correlating with a higher 5-year RR (67%) than other etiologies. CONCLUSION Our study introduces tumor marker clustering as a novel analytical approach, providing a nuanced understanding of AFP and DCP's combined utility in predicting prognosis and recurrence. Our findings highlight the independent and complementary roles of these biomarkers, particularly in NBNC-related HCC and in cases with impaired liver function. AFP and DCP remain crucial tools for recurrence risk assessment, guiding personalized management strategies, such as surveillance, neoadjuvant therapies, and tailored postoperative interventions.
Collapse
Affiliation(s)
- Yoshitaka Saegusa
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuki Imaoka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; Division of Abdominal Transplant, Stanford University, Stanford, CA, United States
| | - Masahiro Ohira
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan.
| | - Tsuyoshi Kobayashi
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Naruhiko Honmyo
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; Department of Surgery, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Michinori Hamaoka
- Department of Gastroenterological, Breast and Transplant Surgery, Hiroshima Prefectural Hospital, Hiroshima, Japan
| | - Takashi Onoe
- Department of Surgery, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure City, Japan
| | - Daisuke Takei
- Department of Surgery, Onomichi General Hospital, Onomichi City, Japan
| | - Koichi Oishi
- Department of Surgery, Chugoku Rosai Hospital, Kure City, Japan
| | - Tomoyuki Abe
- Department of Surgery, National Hospital Organization, Higashihiroshima Medical Center, Higashihiroshima, Japan
| | - Toshihiro Nakayama
- Division of Abdominal Transplant, Stanford University, Stanford, CA, United States
| | - Miho Akabane
- Division of Abdominal Transplant, Stanford University, Stanford, CA, United States
| | - Kazunari Sasaki
- Division of Abdominal Transplant, Stanford University, Stanford, CA, United States
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
45
|
Zhang L, Zhang F, Xiao CJ, Shu YF, Li Z, Wang J, Tang WJ. Impact of a Quantitative Early Activity Program on Gastrointestinal Function Following Laparoscopic Pancreaticoduodenectomy: A Single-Center Retrospective Analysis. Surg Laparosc Endosc Percutan Tech 2025; 35:e1357. [PMID: 39895546 DOI: 10.1097/sle.0000000000001357] [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: 09/03/2024] [Accepted: 01/07/2025] [Indexed: 02/04/2025]
Abstract
OBJECTIVE The objective of this study is to assess the impact of a quantitative early activity program, integrated into the Enhanced Recovery After Surgery (ERAS) protocol on gastrointestinal function in patients undergoing laparoscopic pancreatoduodenectomy (LPD). METHODS Perioperative data from 203 patients who underwent LPD at the Department of Comprehensive Treatment of Pancreatic Cancer, Fudan University Shanghai Cancer Center, between January 2021 and December 2022 were analyzed retrospectively. The patients were categorized into 2 groups based on their nursing plans. Group A received the standard perioperative ERAS nursing plan, while group B followed the ERAS plan supplemented with a quantitative early activity program. We assessed postoperative outcomes including bowel sound recovery time, time to first anal exhaust, time to first mobilization, activity compliance 1-week postsurgery, and incidence of postoperative breakthrough pain. RESULT We compared several postoperative metrics between group A and group B. Specifically, the time to bowel sound recovery was 62.39±17.89 hours in group A versus 56.45±22.85 hours in group B. The time to first anal exhaust was 78.88±71.99 hours in group A compared with 63.62±24.73 hours in group B. The time to first mobilization was 56.98±18.66 hours in group A versus 49.85±20.48 hours in group B. In addition, activity compliance 1-week postsurgery and the incidence of postoperative breakthrough pain (1.55±2.01 times in group A vs. 0.94±1.16 times in group B) were also compared. All these differences were statistically significant ( P <0.05). Conversely, the incidence of postoperative complications and the length of hospital stay (11.20±5.69 d in group A vs. 12.47±6.67 d in group B) did not reveal any significant differences ( P >0.05). CONCLUSION The quantitative early activity program for LPD, based on the ERAS protocol, enhances the adherence to postoperative activity and decreases the incidence of breakthrough pain, thereby facilitating gastrointestinal function recovery in patients. This approach merits clinical adoption.
Collapse
Affiliation(s)
- Ling Zhang
- Departments of Nursing Administration, Fudan University Shanghai Cancer Center
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Cente
| | - Fan Zhang
- Departments of Nursing Administration, Fudan University Shanghai Cancer Center
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Cente
| | - Chen-Jie Xiao
- Departments of Nursing Administration, Fudan University Shanghai Cancer Center
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Cente
| | - Yue-Fen Shu
- Departments of Nursing Administration, Fudan University Shanghai Cancer Center
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Cente
| | - Zheng Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Cente
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai China
| | - Jun Wang
- Departments of Nursing Administration, Fudan University Shanghai Cancer Center
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Cente
| | - Wen-Jie Tang
- Departments of Nursing Administration, Fudan University Shanghai Cancer Center
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Cente
| |
Collapse
|
46
|
Su H, Zhou X, Lin G, Luo C, Meng W, Lv C, Chen Y, Wen Z, Li X, Wu Y, Xiao C, Yang J, Lu J, Luo X, Chen Y, Tam PKH, Li C, Sun H, Pan X. Deciphering the Oncogenic Landscape of Hepatocytes Through Integrated Single-Nucleus and Bulk RNA-Seq of Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412944. [PMID: 39960344 PMCID: PMC11984907 DOI: 10.1002/advs.202412944] [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: 10/14/2024] [Revised: 01/01/2025] [Indexed: 04/12/2025]
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related mortality, while the hepatocyte mechanisms driving oncogenesis remains poorly understood. In this study, single-nucleus RNA sequencing of samples from 22 HCC patients revealed 10 distinct hepatocyte subtypes, including beneficial Hep0, predominantly malignant Hep2, and immunosuppressive Hep9. These subtypes were strongly associated with patient prognosis, confirmed in TCGA-LIHC and Fudan HCC cohorts through hepatocyte composition deconvolution. A quantile-based scoring method is developed to integrate data from 29 public HCC datasets, creating a Quantile Distribution Model (QDM) with excellent diagnostic accuracy (Area Under the Curve, AUC = 0.968-0.982). QDM was employed to screen potential biomarkers, revealing that PDE7B functions as a key gene whose suppression promotes HCC progression. Guided by the genes specific to Hep0/2/9 subtypes, HCC is categorized into metabolic, inflammatory, and matrix classes, which are distinguishable in gene mutation frequencies, survival times, enriched pathways, and immune infiltration. Meanwhile, the sensitive drugs of the three HCC classes are identified, namely ouabain, teniposide, and TG-101348. This study presents the largest single-cell hepatocyte dataset to date, offering transformative insights into hepatocarcinogenesis and a comprehensive framework for advancing HCC diagnostics, prognostics, and personalized treatment strategies.
Collapse
Affiliation(s)
- Huanhou Su
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Xuewen Zhou
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Guanchuan Lin
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Chaochao Luo
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
- College of Life SciencesShihezi UniversityShiheziXinjiang832003China
| | - Wei Meng
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Cui Lv
- Clinical Biobank CenterMicrobiome Medicine CenterDepartment of Laboratory MedicineGuangdong Provincial Clinical Research Center for Laboratory MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Yuting Chen
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Zebin Wen
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Xu Li
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Yongzhang Wu
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Changtai Xiao
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Jian Yang
- Department of Hepatobiliary Surgery IGeneral Surgery Center and Guangdong Provincial Clinical and Engineering Center of Digital MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Jiameng Lu
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Xingguang Luo
- Department of PsychiatryYale University School of MedicineNew HavenCT06510USA
| | - Yan Chen
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Paul KH Tam
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Chuanjiang Li
- Division of Hepatobiliopancreatic SurgeryDepartment of General SurgeryNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Haitao Sun
- Clinical Biobank CenterMicrobiome Medicine CenterDepartment of Laboratory MedicineGuangdong Provincial Clinical Research Center for Laboratory MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Xinghua Pan
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
- Key Laboratory of Infectious Diseases Research in South China (China Ministry Education)Southern Medical UniversityGuangzhouGuangdong510515China
| |
Collapse
|
47
|
Rangwala HS, Fatima H, Ali M, Rangwala BS. Comparing Safety and Efficacy: Laparoscopic vs. Open Pancreaticoduodenectomy for Pancreatic Ductal Adenocarcinoma: A Meta-analysis of Randomized Control Trials. Indian J Surg Oncol 2025; 16:432-442. [PMID: 40337026 PMCID: PMC12052625 DOI: 10.1007/s13193-024-02132-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 11/11/2024] [Indexed: 05/09/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has an estimated 5-year survival rate of 11% and remains a formidable challenge. Despite evolving pancreaticoduodenectomy techniques, the pinnacle status of laparoscopic surgery persists. Technological advancements since Gagner and Pomp's 1994 breakthrough have expanded its reach, yet it remains complex. Although safety reports exist, large-scale studies establishing the viability of laparoscopic surgery for PDAC are scarce. This study aimed to provide an up-to-date meta-analysis, scrutinizing all relevant trials, especially recently published ones, to discern differences between laparoscopic and open pancreaticoduodenectomy procedures. This meta-analysis, following the PRISMA guidelines, systematically searched the PubMed and Cochrane Library databases for randomized controlled trials on pancreaticoduodenectomy up to November 25, 2023. Data extraction, quality assessment, and statistical analysis, including primary and secondary outcomes, were conducted using rigorous methodology. The study employed Review Manager 5.4 for analysis, with significance set at P < 0.05. This meta-analysis, comprising five randomized controlled trials (laparoscopic, 605; open, 601; total, 1206), utilized the Cochrane Risk of Bias 2 tool, revealing minimal bias. Primary outcome analysis indicated a shorter hospital stay with laparoscopic pancreaticoduodenectomy (S.M.D. = - 0.18, 95% CI - 0.63 - 0.28, P = 0.45) and reduced blood loss (S.M.D. = - 1.96, 95% CI - 3.05, 0.88, P = 0.0004), but increased operative time (S.M.D. = 1.74, 95% CI 0.95 to 2.53, P < 0.0001). The secondary outcomes showed no significant differences in morbidity, mortality, resection, fistula, gastric emptying, hemorrhage, bile leak, reoperation, readmission, and surgical site infection between the two procedures. Laparoscopic pancreaticoduodenectomy (LPD) offers marginal benefits over open pancreaticoduodenectomy (OPD), including reduced blood loss and shorter hospital stays. However, LPD requires a significantly longer operative time, and no substantial differences have been observed in terms of mortality or other complications. Additional studies with lower heterogeneity are needed to thoroughly evaluate the comparative effectiveness and safety of LPD and OPD.
Collapse
Affiliation(s)
- Hussain Sohail Rangwala
- Department of Medicine, Jinnah Sindh Medical University, Iqbal, Shaheed Rd, Karachi, Pakistan
| | - Hareer Fatima
- Department of Medicine, Jinnah Sindh Medical University, Iqbal, Shaheed Rd, Karachi, Pakistan
| | - Mirha Ali
- Department of Medicine, Jinnah Sindh Medical University, Iqbal, Shaheed Rd, Karachi, Pakistan
| | | |
Collapse
|
48
|
Joshi P, Nascimento HSD, Kang SY, Lee M, Vanga MG, Lee SH, Ku B, Miranda MDS, Lee MY. Dynamic Culture of Bioprinted Liver Tumor Spheroids in a Pillar/Perfusion Plate for Predictive Screening of Anticancer Drugs. Biotechnol Bioeng 2025; 122:995-1009. [PMID: 39821523 DOI: 10.1002/bit.28924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/13/2024] [Accepted: 01/02/2025] [Indexed: 01/19/2025]
Abstract
Recent advancements in three-dimensional (3D) cell culture technologies, such as cell spheroids, organoids, and 3D bioprinted tissue constructs, have significantly improved the physiological relevance of in vitro models. These models better mimic tissue structure and function, closely emulating in vivo characteristics and enhancing phenotypic analysis, critical for basic research and drug screening in personalized cancer therapy. Despite their potential, current 3D cell culture platforms face technical challenges, which include user-unfriendliness in long-term dynamic cell culture, incompatibility with rapid cell encapsulation in biomimetic hydrogels, and low throughput for compound screening. To address these issues, we developed a 144-pillar plate with sidewalls and slits (144PillarPlate) and a complementary 144-perfusion plate with perfusion wells and reservoirs (144PerfusionPlate) for dynamic 3D cell culture and predictive compound screening. To accelerate biomimetic tissue formation, small Hep3B liver tumor spheroids suspended in alginate were printed and encapsulated on the 144PillarPlate rapidly by using microsolenoid valve-driven 3D bioprinting technology. The microarray bioprinting technology enabled precise and rapid loading of small spheroids in alginate on the pillar plate, facilitating reproducible and scalable formation of large tumor spheroids with minimal manual intervention. The bioprinted Hep3B spheroids on the 144PillarPlate were dynamically cultured in the 144PerfusionPlate and tested with anticancer drugs to measure drug effectiveness and determine the concentration required to inhibit 50% of the cell viability (IC50 value). The perfusion plate enabled the convenient dynamic culture of tumor spheroids and facilitated the dynamic testing of anticancer drugs with increased sensitivity. It is envisioned that the integration of microarray bioprinting of tumor spheroids onto the pillar plate, along with dynamic 3D cell culture in the perfusion plate, could more accurately replicate tumor microenvironments. This advancement has the potential to enhance the predictive drug screening process in personalized cancer therapy significantly.
Collapse
Affiliation(s)
- Pranav Joshi
- Bioprinting Laboratories Inc., Dallas, Texas, USA
| | - Hamilton Silva do Nascimento
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA
- Institute of Veterinary Medicine, Federal University of Para, Castanhal, Brazil
| | - Soo-Yeon Kang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA
| | - Minseong Lee
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA
| | | | | | - Bosung Ku
- MBD Co. Ltd., Suwon, Republic of Korea
| | | | - Moo-Yeal Lee
- Bioprinting Laboratories Inc., Dallas, Texas, USA
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA
| |
Collapse
|
49
|
Ni X, Wei Z, Peng Y, Zheng L, Shang J, Liu F, Li Y, Liu J. Triclosan exposure induces liver fibrosis in mice: The heterogeneous nuclear ribonucleoprotein A1/pyruvate kinase M2 axis drives hepatic stellate cell activation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 294:118113. [PMID: 40157328 DOI: 10.1016/j.ecoenv.2025.118113] [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: 11/07/2024] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Triclosan (TCS) is an effective broad-spectrum antibacterial agent. TCS possesses a stable structure, can easily accumulate in the environment, and may have numerous negative impacts on human health. One organ particularly susceptible to TCS damage is the liver; however, the molecular mechanisms underlying TCS-induced liver damage remain unclear. A long-term TCS exposure model was established in C57BL/6 mice through maternal administration from gestation to postnatal 8-week-old. The offspring were randomly assigned to three groups (0, 50, and 100 mg/kg TCS) with six animals per group, ensuring an equal gender distribution (3 males and 3 females). The results showed that TCS-exposed mice exhibited serum aspartate aminotransferase, alanine aminotransferase, and alkaline phosphatase enzyme activities increased by 1.5-2 times when compared with vehicle-treated mice, along with features of liver fibrosis. In the LX-2 cell line, used as an in vitro model, TCS promoted proliferation and migration and induced the activation of hepatic stellate cells (HSCs). The level of pyruvate kinase M2 (PKM2) dimer increased by 200 % in LX-2 cells treated with TCS. PKM2 dimer overexpression stimulated HSC activation, whereas treatment with TEPP-46 (a PKM2 dimer inhibitor) significantly decreased the activation process. The expression of heterogeneous ribonucleoprotein particle A1 (hnRNPA1) was upregulated in the TCS treatment group and promoted the PKM2 expression. Moreover, disruption of the hnRNPA1/PKM2 axis reduced HSC proliferation and migration activated by TCS. Overall, our findings highlighted that TCS could cause liver fibrosis by stimulating the proliferation and migration of HSCs activated via the hnRNPA1/PKM2 axis, providing promising treatment options for TCS-related liver damage.
Collapse
Affiliation(s)
- Xiao Ni
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Ziyun Wei
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Yuxuan Peng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Linlin Zheng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Jianing Shang
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Fu Liu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Yunwei Li
- Department of Anorectal Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China.
| | - Jieyu Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, PR China; Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China.
| |
Collapse
|
50
|
Tan J, Li F, Zhang X, Zhu H, Liu J, Wu T, Zhang Y, Zhang D, Geng Y, Shen Y. Extracts from petal of the Crocus sativus (saffron) possesses detoxification effects on acetaminophen induced liver injury by inhibiting hepatocyte apoptosis via regulating Nrf2/HO-1 signaling. Fitoterapia 2025; 182:106452. [PMID: 39993543 DOI: 10.1016/j.fitote.2025.106452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 02/10/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
The purpose of this study was to investigate the detoxification effect of extracts from the petal of Crocus sativus L. (PCSE) on acetaminophen (APAP) induced liver injury in mice and its related mechanisms. LC-MS/MS analysis was used to identify the main components in PCSE, and an APAP-induced acute liver injury model in mice was constructed to evaluate the detoxification effect of PCSE. Liver tissue H&E staining, liver function indexes including ALT and AST, pro-inflammatory cytokine including TNF-α and IL-6, as well as hepatic tissue oxidative stress levels were examined. In addition, in vitro APAP induced cell was also prepared, apoptosis levels were detected by AO/EB staining, ROS fluorescence intensity was analyzed as well as the expression levels of apoptosis-related proteins and Nrf2/HO-1 pathway-related proteins were detected by western blot, to investigate the mechanism of PCSE's action in ameliorating liver injury. The results showed that PCSE can improve the survival rate of APAP induced mice, decrease ALT, AST, TNF-α and IL-6 levels, and ameliorate the liver injury induced by APAP. Furthermore, the mechanism research suggested PCSE attenuated oxidative stress and apoptosis in APAP-induced liver cells, as well as activated the Nrf2/HO-1 signaling. In summary, PCSE possesses potential detoxification effects on APAP induced liver injury by inhibiting hepatocyte apoptosis via regulating Nrf2/HO-1 signaling, which provides more possibilities for the drug selection for the treatment of liver injury in clinical practice.
Collapse
Affiliation(s)
- Jin Tan
- Gooddoctor Pharmaceutical Group Co., Ltd., Chengdu 610073, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Fangqiong Li
- Gooddoctor Pharmaceutical Group Co., Ltd., Chengdu 610073, China
| | - Xin Zhang
- Gooddoctor Pharmaceutical Group Co., Ltd., Chengdu 610073, China
| | - Hongrui Zhu
- Gooddoctor Pharmaceutical Group Co., Ltd., Chengdu 610073, China
| | - Jin Liu
- Gooddoctor Pharmaceutical Group Co., Ltd., Chengdu 610073, China
| | - Taoqing Wu
- Gooddoctor Pharmaceutical Group Co., Ltd., Chengdu 610073, China
| | - Yang Zhang
- Gooddoctor Pharmaceutical Group Co., Ltd., Chengdu 610073, China
| | - Dingkun Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuefei Geng
- Gooddoctor Pharmaceutical Group Co., Ltd., Chengdu 610073, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yongmei Shen
- Gooddoctor Pharmaceutical Group Co., Ltd., Chengdu 610073, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|