1
|
Poudineh M, Mohammadyari F, Parsamanesh N, Jamialahmadi T, Kesharwani P, Sahebkar A. Cell and gene therapeutic approaches in non-alcoholic fatty liver disease. Gene 2025; 956:149466. [PMID: 40189164 DOI: 10.1016/j.gene.2025.149466] [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: 01/06/2025] [Revised: 03/14/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) refers to a range of conditions marked by the buildup of triglycerides in liver cells, accompanied by inflammation, which contributes to liver damage, clinical symptoms, and histopathological alterations. Multiple molecular pathways contribute to NAFLD pathogenesis, including immune dysregulation, endoplasmic reticulum stress, and tissue injury. Both the innate and adaptive immune systems play crucial roles in disease progression, with intricate crosstalk between liver and immune cells driving NAFLD development. Among emerging therapeutic strategies, cell and gene-based therapies have shown promise. This study reviews the pathophysiological mechanisms of NAFLD and explores the therapeutic potential of cell-based interventions, highlighting their immunomodulatory effects, inhibition of hepatic stellate cells, promotion of hepatocyte regeneration, and potential for hepatocyte differentiation. Additionally, we examine gene delivery vectors designed to target NAFLD, focusing on their role in engineering hepatocytes through gene addition or editing to enhance therapeutic efficacy.
Collapse
Affiliation(s)
| | | | - Negin Parsamanesh
- Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Tananz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact and Outcome, Chitkara University, Rajpura 140417, Punjab, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Liu Z, Han J, Su S, Zeng Q, Wu Z, Yuan J, Yang J. Histone lactylation facilitates MCM7 expression to maintain stemness and radio-resistance in hepatocellular carcinoma. Biochem Pharmacol 2025; 236:116887. [PMID: 40118288 DOI: 10.1016/j.bcp.2025.116887] [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: 12/24/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Cancer stem cells (CSCs) play an essential role in tumor initiation and therapy resistance. Histone lactylation as a novel epigenetic modification could regulate the gene transcription process during tumor progression. Nevertheless, researches have not well examined its role in maintaining CSC properties. Our study identified Minichromosome maintenance complex component 7 (MCM7) as a candidate gene in Hepatocellular carcinoma (HCC) with diagnostic and prognostic values, and Real-time quantitative PCR (qRT-PCR), Western blot (WB), and Immunohistochemistry (IHC) assays ascertained its obviously higher expressions in HCC cells and tissues. Ectopic of MCM7 could increase the expression of CSC-related genes and enhance spheroid both in size and in number. Suppression of MCM7 could strengthen the efficacy of radiotherapy verified by Cell counting kit-8 (CCK-8) and colony formation assays. The subcutaneous xenograft model indicated that suppression of MCM7 could inhibit CSC properties and the efficacy of radiotherapy in vivo. Mechanistically, histone lactylation could facilitate MCM7 expression, and both messenger RNA (mRNA) and protein level of MCM7 expression presented an obvious decrease due to 2-DG (glycolysis inhibitor) treatment and an obvious increase due to Rotenone (glycolysis activator) treatment. Rescue experiments verified that histone lactylation was necessary for MCM7 to promote CSC properties and radio-resistance in HCC. Arsenic trioxide (ATO) targeting MCM7 could inhibit the CSC phenotypes and enhance the efficacy of radiotherapy in vivo and in vitro. Collectively, histone lactylation could transcriptionally activate MCM7 to accelerate proliferation and radio-resistance through enhancing CSC properties. ATO targeting MCM7 could inhibit CSCs phenotypes and synergistically increase the efficacy of radiation therapy.
Collapse
Affiliation(s)
- Zijian Liu
- Laboratory of Liquid Biopsy and Single Cell Research, Department of Radiation Oncology and Department of Head and Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiaqi Han
- Laboratory of Liquid Biopsy and Single Cell Research, Department of Radiation Oncology and Department of Head and Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shitong Su
- Laboratory of Liquid Biopsy and Single Cell Research, Department of Radiation Oncology and Department of Head and Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qiwen Zeng
- Institute of Organ Transplantation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenru Wu
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingsheng Yuan
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Institute of Organ Transplantation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China.
| | - Jian Yang
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Institute of Organ Transplantation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
3
|
Xu Y, Li X, Cui M, Pan T, Zheng S, Shang Z, Yin D, Xu Y, Yang Z, Wang J, Song X. Fufang-Biejia-Ruangan tablet targeting both cancer-associated fibroblasts and tumor cells by HIPPO-PI3K/AKT cascades in intrahepatic cholangiocarcinoma treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156690. [PMID: 40215817 DOI: 10.1016/j.phymed.2025.156690] [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: 01/03/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (ICC) ranks second among primary liver cancers in terms of prevalence, characterized by poor prognosis and scarce therapeutic interventions. Fufang-Biejia-Ruangan tablet (BJRG), a traditional Chinese herbal remedy, has been widely used for liver diseases. However, its therapeutic efficacy and underlying mechanisms in ICC remain poorly understood. AIM OF THE STUDY This study aims to systematically investigate the anti-ICC effects of BJRG, focusing on tumor progression and microenvironment modulation, through experimental and transcriptomic analyses. METHODS The chemical composition of BJRG was analyzed employing ultra-performance liquid chromatography-mass spectrometry (UPLC-MS). In vitro assays were performed with QBC939 and LX-2 cell lines. Two primary ICC models (AKT/YAP and sgP53/KRAS) were established via hydrodynamic tail-vein injection of corresponding plasmids. A co-culture system for subcutaneous tumor formation was developed using cancer-associated fibroblasts (CAFs) derived from the AKT/YAP model and primary tumor cells derived from the sgP53/KRAS model. RESULTS UPLC-MS analysis identified 1091 chemical components, primarily terpenoids, sugars, glycosides, and phenylpropanoids. The therapeutic efficacy of BJRG was evaluated for the treatment of ICC. BJRG treatment slowed down the growth of both human ICC cell lines and AKT/YAP ICC mouse model. Mechanistically, BJRG inhibited HIPPO-PI3K/AKT signaling pathway in ICC tumor cells. Importantly, BJRG significantly inhibited the growth of CAFs via HIPPO-PI3K/AKT cascades. Of note, co-culture CAFs with ICC cell lines substantially sensitized the efficacy of BJRG in sgP53/KRAS syngeneic tumor model. Furthermore, BJRG therapy not only affected CAFs but also induced alterations in vascular structures and hypoxic conditions within lesions in the AKT/YAP model. This intervention promoted the infiltration of T lymphocytes and macrophages into the tumor microenvironment, which may further augment the anti-proliferative effects of BJRG by enhancing the immune response within ICC tumor tissues. CONCLUSION Our research demonstrates BJRG's anti-ICC efficacy via diverse pathways, including the suppression of tumor cell proliferation, regulation of CAFs activity, and promotion of immune cell infiltration. These findings underscore BJRG as a promising therapeutic candidate for ICC, offering novel mechanistic insights and highlighting its potential for clinical translation.
Collapse
Affiliation(s)
- Yanyu Xu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian 350122, China; Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Xiang Li
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Miao Cui
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Tao Pan
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Shuwen Zheng
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian 350122, China; Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Zimeng Shang
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; Center of Integrative Medicine, Beijing Ditan Hospital, Affiliated to Capital Medical University, Beijing 100015, China
| | - Donghao Yin
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; Beijing You'an Hospital, Affiliated to Capital Medical University, Beijing, 100069, China
| | - Yang Xu
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Zhiyun Yang
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; Center of Integrative Medicine, Beijing Ditan Hospital, Affiliated to Capital Medical University, Beijing 100015, China
| | - Jiabo Wang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian 350122, China; Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China.
| | - Xinhua Song
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
4
|
Viola H, Chen LH, Jo S, Washington K, Selva C, Li A, Feng D, Giacalone V, Stephenson ST, Cottrill K, Mohammad A, Williams E, Qu X, Lam W, Ng NL, Fitzpatrick A, Grunwell J, Tirouvanziam R, Takayama S. High-throughput quantitation of human neutrophil recruitment and functional responses in an air-blood barrier array. APL Bioeng 2025; 9:026110. [PMID: 40290728 PMCID: PMC12033047 DOI: 10.1063/5.0220367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 11/19/2024] [Indexed: 04/30/2025] Open
Abstract
Dysregulated neutrophil recruitment drives many pulmonary diseases, but most preclinical screening methods are unsuited to evaluate pulmonary neutrophilia, limiting progress toward therapeutics. Namely, high-throughput therapeutic assays typically exclude critical neutrophilic pathophysiology, including blood-to-lung recruitment, dysfunctional activation, and resulting impacts on the air-blood barrier. To meet the conflicting demands of physiological complexity and high throughput, we developed an assay of 96-well leukocyte recruitment in an air-blood barrier array (L-ABBA-96) that enables in vivo-like neutrophil recruitment compatible with downstream phenotyping by automated flow cytometry. We modeled acute respiratory distress syndrome (ARDS) with neutrophil recruitment to 20 ng/mL epithelial-side interleukin 8 and found a dose-dependent reduction in recruitment with physiologic doses of baricitinib, a JAK1/2 inhibitor recently Food and Drug Administration-approved for severe Coronavirus Disease 2019 ARDS. Additionally, neutrophil recruitment to patient-derived cystic fibrosis sputum supernatant induced disease-mimetic recruitment and activation of healthy donor neutrophils and upregulated endothelial e-selectin. Compared to 24-well assays, the L-ABBA-96 reduces required patient sample volumes by 25 times per well and quadruples throughput per plate. Compared to microfluidic assays, the L-ABBA-96 recruits two orders of magnitude more neutrophils per well, enabling downstream flow cytometry and other standard biochemical assays. This novel pairing of high-throughput in vitro modeling of organ-level lung function with parallel high-throughput leukocyte phenotyping substantially advances opportunities for pathophysiological studies, personalized medicine, and drug testing applications.
Collapse
Affiliation(s)
| | - Liang-Hsin Chen
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Seongbin Jo
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Kendra Washington
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Cauviya Selva
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Andrea Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Daniel Feng
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | | | | | | | | | - Evelyn Williams
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Xianggui Qu
- Department of Mathematics and Statistics, Oakland University, Rochester, Michigan 48309, USA
| | | | | | | | - Jocelyn Grunwell
- Department of Pediatrics, Division of Critical Care Medicine, Emory University School of Medicine and Children's Healthcare of Atlanta at Arthur M. Blank Hospital, Atlanta, Georgia 30322, USA
| | | | | |
Collapse
|
5
|
Ma L, Wang W, Gu L, Wang L. cPLA 2α on the influence of Th17 and its role in the formation of liver fibrosis. Cytotechnology 2025; 77:87. [PMID: 40206205 PMCID: PMC11977053 DOI: 10.1007/s10616-025-00750-6] [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: 03/29/2025] [Indexed: 04/11/2025] Open
Abstract
This study primarily investigated the mechanism and pathways of the cPLA2α signaling pathway on Th17-mediated HSC activation and liver fibrosis, providing insights for clinical strategies to target HSC activation and delay the rapid progression of liver fibrosis. In vitro and in vivo model were established, and different concentrations of the cPLA2α inhibitor AACOCF3 were administered respectively for intervention. The expression of IL- 17 was detected by ELISA, and the expression of cPLA2α protein and HSC activation protein α-SMA index were detected by Western blot and immunofluorescence. In addition, observe the changes in the degree of liver fibrosis in mice through the pathological staining of mouse livers. In an in vitro system, Th17 could induce HSC activation. And after intervention, the results showed that the inhibitor could inhibit Th17 activation of HSC. Next, in an in vivo model, Th17 could also induce HSC activation. And after intervention, the results showed that the inhibitor could also inhibit HSC activation by Th17. Observation under liver pathological staining showed that the inflammation and staining were significantly reduced in the intervention group, suggesting a therapeutic effect of AACOCF3. Using in vitro and in vivo approaches, these data suggest that Th17 cells can promote the activation and proliferation of HSCs, which further exerts a role in promoting liver fibrosis. These data also suggest that the cPLA2α pathway may be involved in the activation of HSCs by Th17 cells and induce liver fibrosis mechanisms.
Collapse
Affiliation(s)
- Lina Ma
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014 Shandong China
- Shandong First Medical University, Jinan, 250117 Shandong China
| | - Wei Wang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014 Shandong China
- Shandong First Medical University, Jinan, 250117 Shandong China
| | - Limin Gu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014 Shandong China
- Shandong First Medical University, Jinan, 250117 Shandong China
| | - Liyun Wang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014 Shandong China
| |
Collapse
|
6
|
Xu ZY, Wang M, Shi JY, Liu Y, Yu C, Zhang XY, Zhang CW, He QF, Pan C, Zhou J, Xiao H, Cao HY, Ma Y. Engineering a dynamic extracellular matrix using thrombospondin-1 to propel hepatocyte organoids reprogramming and improve mouse liver regeneration post-transplantation. Mater Today Bio 2025; 32:101700. [PMID: 40225139 PMCID: PMC11986605 DOI: 10.1016/j.mtbio.2025.101700] [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: 01/14/2025] [Revised: 03/09/2025] [Accepted: 03/22/2025] [Indexed: 04/15/2025] Open
Abstract
Hepatocyte organoids (HOs) hold significant potential for constructing bioartificial liver construction, toxicology research, and liver failure therapies. However, challenges such as difficulties in induced pluripotent stem cells (iPSCs) harvest and differentiation, safety concerns of tumor-derived matrices, and limited primary cell regulation hinder clinical applications. In this study, we developed a non-tumor-derived decellularized extracellular matrix (dECM) system with tunable mechanical properties and viscoelasticity to enhance stem cell proliferation and organoid functionality using thrombospondin-1 (THBS1). Nanoindentation and transcriptomic analysis revealed that THBS1 mediates adaptation and remodeling between organoids and ECM proteins, exhibiting native tissue-like viscoelasticity and up-regulated reprogramming transcriptional factors KLF4 and SOX2 via the YAP/TAZ pathway. Transplanting HOs presenting reprogramming effects into a 70 % hepatectomy model demonstrated improved liver regeneration, underscoring the potential of the THBS1-based dynamic ECM system in organoids manipulation and liver regeneration.
Collapse
Affiliation(s)
- Zi-Yan Xu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Min Wang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jing-Yan Shi
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ye Liu
- School of Medicine, Southeast University, Nanjing, China
| | - Chao Yu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xin-Yi Zhang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Chen-Wei Zhang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qi-Feng He
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Chao Pan
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jin Zhou
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hua Xiao
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hong-Yong Cao
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yong Ma
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
7
|
Liang G, Ma Y, Deng P, Li S, He C, He H, Liu H, Fan Y, Li Z. Role of cell-based therapies in digestive disorders: Obstacles and opportunities. Regen Ther 2025; 29:1-18. [PMID: 40124469 PMCID: PMC11925584 DOI: 10.1016/j.reth.2025.02.009] [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: 12/22/2024] [Revised: 02/01/2025] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
Stem cell-based therapies have emerged as a promising frontier in the treatment of gastrointestinal disorders, offering potential solutions for challenges posed by conventional treatments. This review comprehensively examines recent advancements in cell-based therapeutic strategies, particularly focusing on stem cell applications, immunotherapy, and cellular therapies for digestive diseases. It highlights the successful differentiation of enteric neural progenitors from pluripotent stem cells and their application in animal models, such as Hirschsprung disease. Furthermore, the review evaluates clinical trials and experimental studies demonstrating the potential of stem cells in regenerating damaged tissues, modulating immune responses, and promoting healing in conditions like Crohn's disease and liver failure. By addressing challenges, such as scalability, immunogenicity, and ethical considerations, the review underscores the translational opportunities and obstacles in realizing the clinical potential of these therapies. Concluding with an emphasis on future directions, the study provides insights into optimizing therapeutic efficacy and fostering innovations in personalized medicine for digestive disorders.
Collapse
Affiliation(s)
- Guodong Liang
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Yuehan Ma
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Ping Deng
- Medical Department, Jilin Cancer Hospital, Changchun 130012, China
| | - Shufeng Li
- First Department of Gynecological Tumor, Jilin Cancer Hospital, Changchun 130012, China
| | - Chunyan He
- Department of Anaesthesia, Jilin Cancer Hospital, Changchun 130012, China
| | - Haihang He
- Department of Otorhinolaryngology, Oral Maxillofacial, Head and Neck, Jilin Cancer Hospital, Changchun 130012, China
| | - Hairui Liu
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Yunda Fan
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Ze Li
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| |
Collapse
|
8
|
Nater M, Brügger M, Cecconi V, Pereira P, Forni G, Köksal H, Dimakou D, Herbst M, Calvanese AL, Lucchiari G, Schneider C, Valenta T, van den Broek M. Hepatic iNKT cells facilitate colorectal cancer metastasis by inducing a fibrotic niche in the liver. iScience 2025; 28:112364. [PMID: 40292307 PMCID: PMC12032931 DOI: 10.1016/j.isci.2025.112364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 03/05/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
The liver is an important metastatic organ that contains many innate immune cells, yet little is known about their role in anti-metastatic defense. We investigated how invariant natural killer T (iNKT) cells influence colorectal cancer-derived liver metastasis using different models in immunocompetent mice. We found that hepatic iNKT cells promote metastasis by creating a supportive niche for disseminated cancer cells. Mechanistically, iNKT cells respond to disseminating cancer cells by producing the fibrogenic cytokines interleukin-4 (IL-4) and IL-13 in a T cell receptor-independent manner. Selective abrogation of IL-4 and IL-13 sensing in hepatic stellate cells prevented their transdifferentiation into extracellular matrix-producing myofibroblasts, which hindered metastatic outgrowth of disseminated cancer cells. This study highlights a novel tumor-promoting axis driven by iNKT cells in the initial stages of metastasis.
Collapse
Affiliation(s)
- Marc Nater
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Michael Brügger
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Virginia Cecconi
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Paulo Pereira
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Geo Forni
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Hakan Köksal
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Despoina Dimakou
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Michael Herbst
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Giulia Lucchiari
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Tomas Valenta
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
9
|
Attanasio S. Autophagy in cancer and protein conformational disorders. FEBS Lett 2025. [PMID: 40342093 DOI: 10.1002/1873-3468.70061] [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: 01/23/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025]
Abstract
Autophagy is a catabolic process by which cells maintain cellular homeostasis through the degradation of dysfunctional cytoplasmic components, such as toxic misfolded proteins and damaged organelles, within the lysosome. It is a multistep process that is tightly regulated by nutrient, energy, and stress-sensing mechanisms. Autophagy plays a pivotal role in various biological processes, including protein and organelle quality control, defense against pathogen infections, cell metabolism, and immune surveillance. As a result, autophagy dysfunction is linked to a variety of pathological conditions. The role of autophagy in cancer is complex and dynamic. Depending on the context, autophagy can have both tumor-suppressive and pro-tumorigenic effects. In contrast, its role is more clearly defined in protein conformational disorders, where autophagy serves as a mechanism to reduce toxic protein aggregation, thereby improving cellular homeostasis. Because autophagy-based therapies hold promising potential for the treatment of cancer and protein conformational disorders, this review will highlight the latest findings and advancements in these areas.
Collapse
Affiliation(s)
- Sergio Attanasio
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
10
|
Walker SE, Yu K, Burgess S, Echeverri K. Neuronal activation in the axolotl brain promotes tail regeneration. NPJ Regen Med 2025; 10:22. [PMID: 40341072 DOI: 10.1038/s41536-025-00413-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025] Open
Abstract
The axolotl retains a remarkable capacity for regenerative repair and is one of the few vertebrate species capable of regenerating its brain and spinal cord after injury. To date, studies investigating axolotl spinal cord regeneration have placed particular emphasis on understanding how cells immediately adjacent to the injury site respond to damage to promote regenerative repair. How neurons outside of this immediate injury site respond to an injury remains unknown. Here, we identify a population of dpErk+/etv1+ glutamatergic neurons in the axolotl telencephalon that are activated in response to injury and are essential for tail regeneration. Furthermore, these neurons project to the hypothalamus where they upregulate the neuropeptide neurotensin in response to injury. Together, these findings identify a unique population of neurons in the axolotl brain whose activation is necessary for successful tail regeneration, and sheds light on how neurons outside of the immediate injury site respond to an injury.
Collapse
Affiliation(s)
- S E Walker
- Marine Biological Laboratory, Eugene Bell Center for Regenerative Biology, Woods Hole, MA, USA
| | - K Yu
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - S Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - K Echeverri
- Marine Biological Laboratory, Eugene Bell Center for Regenerative Biology, Woods Hole, MA, USA.
| |
Collapse
|
11
|
Fang Y, Chen L, Yuan Y, Zhou S, Fu J, Zhang Q, Zhang N, Huang Y, Li Y, Yuan L, Chen L, Xiang C. Human menstrual blood-derived stem cells secreted ECM1 directly interacts with LRP1α to ameliorate hepatic fibrosis through FoxO1 and mTOR signaling pathway. Stem Cell Res Ther 2025; 16:230. [PMID: 40336034 PMCID: PMC12060366 DOI: 10.1186/s13287-025-04351-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/17/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Human menstrual blood-derived stem cells (MenSCs), a major class of mesenchymal stem cells (MSCs), modulate intercellular signals via paracrine factors. Previous studies found that MenSC-derived secretomes exert protective effects against liver fibrosis. However, the underlying mechanisms of these observations remain unclear. METHODS Extracellular Matrix Protein 1 (ECM1), identified in MenSCs culture medium using mass spectrometry, was employed to stably overexpress ECM1-HA or silence in MenSCs using lentiviral vectors. These genetically engineered cells were either intravenously injected into the carbon tetrachloride (CCl4)-induced liver fibrosis mice or co-cultured with hepatic stellate cells (HSCs)-LX-2. The interaction between ECM1 and low-density lipoprotein receptor-related protein 1α (LRP1α) was confirmed using Co-Immunoprecipitation (Co-ip), Duolink Proximity Ligation Assays (PLA) and pull-down. LRP1 deficient mice were generated via intravenous administration of adeno-associated-virus-8. The downstream molecular mechanisms were characterized by non-target metabolomics and multiplex immunohistochemical staining. RNA sequencing was performed to evaluate the genetic alterations in various genes within the MenSCs. RESULTS MenSC-secreted ECM1 exhibits potential to ameliorate liver fibrosis by inactivating HSCs, improving liver functions, and reducing collagen deposition in both cellular and mouse model of the CCl4-induced liver fibrosis. Mechanistically, a novel interaction was identified that ECM1 directly bound to cell surface receptor LRP1α. Notably, the antifibrotic efficacy of MenSC was negated in LRP1-deficient cells and mice. Moreover, the ECM1-LRP1 axis contributed to the alleviation of liver fibrosis by suppressing AKT/mTOR while activating the FoxO1 signaling pathway, thereby facilitating pyrimidine and purine metabolism. Additionally, ECM1-modified MenSCs regulate the transcription of intrinsic cytokine genes, further mitigating liver fibrosis. CONCLUSIONS These findings highlight an extensive network of ECM1-LRP1 interaction, which serve as a link for providing promising insights into the mechanism of MenSC-based drug development for liver fibrosis. Our study also potentially presents novel avenues for clinical antifibrotic therapy.
Collapse
Affiliation(s)
- Yangxin Fang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Lin Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Yin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Sining Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Jiamin Fu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Qi Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Ning Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Yuqi Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Yifei Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Li Yuan
- Innovative Precision Medicine (IPM) Group, Hangzhou, Zhejiang, 311215, China
| | - Lijun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China.
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China.
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, 100730, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, 250117, China.
| |
Collapse
|
12
|
Tian C, Wang Y, Wang R, Pan L, Xu T. Pharmacological and therapeutic effects of natural products on liver regeneration-a comprehensive research. Chin Med 2025; 20:57. [PMID: 40329344 PMCID: PMC12057117 DOI: 10.1186/s13020-025-01108-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/12/2025] [Indexed: 05/08/2025] Open
Abstract
Liver regeneration (LR) refers to the physiological process by which hepatocytes undergo cellular proliferation to restore the structure and function of the liver following significant hepatocyte loss due to injury or partial hepatectomy (PH). While the liver possesses a remarkable regenerative capacity, this process is tightly regulated to ensure appropriate cessation once homeostasis is reestablished. Various strategies, including technological interventions and pharmacological agents, have been explored to enhance LR. Among these, natural products have emerged as promising candidates for promoting LR. For instance, quercetin, a natural compound, has been shown to enhance LR following PH by maintaining redox homeostasis and stimulating hepatocyte proliferation. However, natural products present certain limitations, such as poor solubility and low bioavailability, which may hinder their clinical application. Modifications in the formulation and mode of administration have demonstrated potential in overcoming these challenges and optimizing their pharmacological effects. Recent advancements in research have further highlighted the growing relevance of natural products, including traditional Chinese medicine (TCM), in the context of LR. Despite this progress, a comprehensive and systematic review of their roles, mechanisms, and therapeutic potential remains lacking. This review aims to bridge this gap by summarizing natural products with demonstrated potential to promote LR. Drawing on data from PubMed, Web of Science, and CNKI databases, it elucidates their pharmacological effects and regulatory mechanisms, providing a valuable reference for future research and clinical application in the field of LR.
Collapse
Affiliation(s)
- Chang Tian
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmaceutical Sciences, Anhui Medical University, Hefei, 230032, China
- Anhui Key Lab of Bioactivity of Natural Products, Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Yuhan Wang
- International Cooperation and Exchange Department, Shanghai General Hospital, 85/86 Wujin Road, Hongkou District, Shanghai, 200434, China
| | - Ran Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmaceutical Sciences, Anhui Medical University, Hefei, 230032, China
- Anhui Key Lab of Bioactivity of Natural Products, Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Linxin Pan
- College of Life Sciences, Anhui Medical University, Hefei, 230032, China.
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmaceutical Sciences, Anhui Medical University, Hefei, 230032, China.
- Anhui Key Lab of Bioactivity of Natural Products, Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
13
|
Zhao Q, Wu J, Feng M, Zhang A, Fu L, Chen J, Li L, Li F, Li T, Jin S, Li S, Yu X. CXCL13 suppresses liver regeneration through the negative regulation of HGF signaling. Cell Death Dis 2025; 16:361. [PMID: 40325003 PMCID: PMC12052986 DOI: 10.1038/s41419-025-07568-2] [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: 11/12/2024] [Revised: 02/24/2025] [Accepted: 03/18/2025] [Indexed: 05/07/2025]
Abstract
Insufficient liver regeneration increases the risk of postoperative liver failure following liver transplantation or partial hepatectomy (PHx). Numerous growth factors and cytokines are related to liver regeneration; however, the underlying mechanisms have not been fully elucidated. In this study, CXCL13 was identified as a key factor delaying liver regeneration after PHx. We observed that CXCL13 expression was upregulated in PHx mice and patients following liver resection. CXCL13 deficiency accelerated liver regeneration, whereas these effects were abolished by recombinant murine CXCL13 administration. Moreover, proteomics analyses indicated that HGF levels in the serum after PHx were significantly greater in Cxcl13-/- mice than in WT mice. Further analysis revealed that CXCL13 deficiency promoted liver regeneration via elevated HGF expression in reparative macrophages and subsequent activated the HGF/c-MET axis in hepatocytes. Additionally, deficiency of macrophage CXCR5, the receptor for CXCL13, augmented liver regeneration and elevated HGF expression after PHx. Mechanistically, CXCL13 inhibited HGF expression in reparative macrophages via CXCR5-mediated AKT/FoxO3a signaling. We further determined that noncanonical NF-κB signaling activation induced CXCL13 expression in hepatic macrophages. Importantly, treatment with CXCL13-neutralizing antibody effectively improved liver regeneration in mice PHx model. Overall, our findings revealed a novel function of CXCL13 in negatively regulating liver regeneration. The underlying mechanism involved CXCL13/CXCR5-mediated FoxO3a signaling, which downregulated HGF expression in reparative macrophages and subsequently attenuated hepatocyte proliferation through inactivating HGF/c-MET signaling. These data suggest that therapeutic targeting of the CXCL13 signaling axis might decrease the risk of postoperative liver failure.
Collapse
Affiliation(s)
- Qun Zhao
- Department of Gastroenterology, Taihe Hospital, School of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
- Laboratory of Inflammation and Molecular Pharmacology, Biomedical Research Institute, Inflammation-Cancer Transformation and Wudang Chinese Medicine Research, Hubei Talent Introduction and Innovation Demonstration Base, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Hubei University of Medicine, Shiyan, China
| | - Jingyi Wu
- Department of Gastroenterology, Taihe Hospital, School of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
- Laboratory of Inflammation and Molecular Pharmacology, Biomedical Research Institute, Inflammation-Cancer Transformation and Wudang Chinese Medicine Research, Hubei Talent Introduction and Innovation Demonstration Base, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Hubei University of Medicine, Shiyan, China
| | - Mengyuan Feng
- Department of Gastroenterology, Taihe Hospital, School of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
- Laboratory of Inflammation and Molecular Pharmacology, Biomedical Research Institute, Inflammation-Cancer Transformation and Wudang Chinese Medicine Research, Hubei Talent Introduction and Innovation Demonstration Base, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Hubei University of Medicine, Shiyan, China
| | - Anjie Zhang
- Department of Gastroenterology, Taihe Hospital, School of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
- Laboratory of Inflammation and Molecular Pharmacology, Biomedical Research Institute, Inflammation-Cancer Transformation and Wudang Chinese Medicine Research, Hubei Talent Introduction and Innovation Demonstration Base, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Hubei University of Medicine, Shiyan, China
| | - Liwei Fu
- Department of Gastroenterology, Taihe Hospital, School of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
- Laboratory of Inflammation and Molecular Pharmacology, Biomedical Research Institute, Inflammation-Cancer Transformation and Wudang Chinese Medicine Research, Hubei Talent Introduction and Innovation Demonstration Base, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Hubei University of Medicine, Shiyan, China
| | - Jinglin Chen
- Department of Clinical Laboratory, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Lian Li
- Department of Clinical Laboratory, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Fangzhou Li
- Laboratory of Inflammation and Molecular Pharmacology, Biomedical Research Institute, Inflammation-Cancer Transformation and Wudang Chinese Medicine Research, Hubei Talent Introduction and Innovation Demonstration Base, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Hubei University of Medicine, Shiyan, China
| | - Tingting Li
- Laboratory of Inflammation and Molecular Pharmacology, Biomedical Research Institute, Inflammation-Cancer Transformation and Wudang Chinese Medicine Research, Hubei Talent Introduction and Innovation Demonstration Base, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Hubei University of Medicine, Shiyan, China
| | - Shu Jin
- Department of Gastroenterology, Taihe Hospital, School of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Shengbao Li
- Department of Gastroenterology, Taihe Hospital, School of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Xianjun Yu
- Department of Gastroenterology, Taihe Hospital, School of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China.
- Laboratory of Inflammation and Molecular Pharmacology, Biomedical Research Institute, Inflammation-Cancer Transformation and Wudang Chinese Medicine Research, Hubei Talent Introduction and Innovation Demonstration Base, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Hubei University of Medicine, Shiyan, China.
| |
Collapse
|
14
|
Sussman JH, Cure HW, Yuan S, Ito K, Asangani IA, Garcia BA, Stanger BZ, Katsuda T. In vivo CRISPR screening reveals epigenetic regulators of hepatobiliary plasticity. Genes Dev 2025; 39:603-616. [PMID: 40169232 PMCID: PMC12047657 DOI: 10.1101/gad.352420.124] [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: 10/29/2024] [Accepted: 02/26/2025] [Indexed: 04/03/2025]
Abstract
Following prolonged liver injury, a small fraction of hepatocytes undergoes reprogramming to become cholangiocytes or biliary epithelial cells (BECs). This physiological process involves chromatin and transcriptional remodeling, but the epigenetic mediators are largely unknown. Here, we exploited a lineage-traced model of liver injury to investigate the role of histone post-translational modification in biliary reprogramming. Using mass spectrometry, we defined the repertoire of histone marks that are globally altered in quantity during reprogramming. Next, applying an in vivo CRISPR screening approach, we identified seven histone-modifying enzymes that alter the efficiency of hepatobiliary reprogramming. Among these, the histone methyltransferase and demethylase Nsd1 and Kdm2a were found to have reciprocal effects on H3K36 methylation that regulated the early and late stages of reprogramming, respectively. Although loss of Nsd1 and Kdm2a affected reprogramming efficiency, cells ultimately acquired the same transcriptomic states. These findings reveal that multiple chromatin regulators exert dynamic and complementary activities to achieve robust cell fate switching, serving as a model for the cell identity changes that occur in various forms of physiological metaplasia or reprogramming.
Collapse
Affiliation(s)
- Jonathan H Sussman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Hector W Cure
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Salina Yuan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Kenji Ito
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Irfan A Asangani
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Benjamin A Garcia
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Ben Z Stanger
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Takeshi Katsuda
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| |
Collapse
|
15
|
Hu Y, Du G, Li C, Wang R, Liu J, Wang Y, Dong J. EGFR-mediated crosstalk between vascular endothelial cells and hepatocytes promotes Piezo1-dependent liver regeneration. Genes Dis 2025; 12:101321. [PMID: 40083329 PMCID: PMC11904541 DOI: 10.1016/j.gendis.2024.101321] [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: 10/04/2023] [Revised: 03/08/2024] [Accepted: 03/31/2024] [Indexed: 03/16/2025] Open
Abstract
Hepatocyte proliferation is essential for recovering liver function after injury. In liver surgery, the mechanical stimulation induced by hemodynamic changes triggers vascular endothelial cells (VECs) to secrete large amounts of cytokines that enhance hepatocyte proliferation and play a pivotal role in liver regeneration (LR). Piezo1, a critical mechanosensory ion channel, can detect and convert mechanical forces into chemical signals, importing external stimuli into cells and triggering downstream biological effects. However, the precise role of Piezo1 in VECs, especially in terms of mediating LR, remains unclear. Here, we report on a potential mechanism by which early changes in hepatic portal hemodynamics activate Piezo1 in VECs to promote hepatocyte proliferation during the process of LR induced by portal vein ligation in rats. In this LR model, hepatocyte proliferation is mainly distributed in zone 1 and zone 2 of liver lobules at 24-48 h after surgery, while only a small number of Ki67-positive hepatocytes were observed in zone 3. Activation of Piezo1 promotes increased secretion of epiregulin and amphiregulin from VECs via the PKC/ERK1/2 axis, further activating epidermal growth factor receptor (EGFR) and ERK1/2 signals in hepatocytes and promoting proliferation. In the liver lobules, the expression of EGFR in hepatocytes of zone 1 and zone 2 is significantly higher than that in zone 3. The EGFR inhibitor gefitinib inhibits LR by suppressing the proliferation of hepatocytes in the middle zone. These data provide a theoretical basis for the regulation of LR through chemical signals mediated by mechanical stimulation.
Collapse
Affiliation(s)
- Yuelei Hu
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun, Jilin 130021, China
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 102218, China
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Guifang Du
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 102218, China
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Chao Li
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 102218, China
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Rui Wang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Juan Liu
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 102218, China
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Yunfang Wang
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 102218, China
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- Institute for Organ Transplant and Bionic Medicine, Tsinghua University, Beijing 100084, China
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Jiahong Dong
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun, Jilin 130021, China
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 102218, China
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- Institute for Organ Transplant and Bionic Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
16
|
Zhan X, Bai Y, Zhu Q, Gao Y, Li F, Bu Q, Zhu Z, Rao Z, Zhou H. Macrophage ATG16L1 promotes liver regeneration after partial hepatectomy. JHEP Rep 2025; 7:101330. [PMID: 40290519 PMCID: PMC12023798 DOI: 10.1016/j.jhepr.2025.101330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 04/30/2025] Open
Abstract
Background & Aims Autophagy plays an important role in liver regeneration. However, most studies are limited to hepatocytes, and the function and mechanism of macrophage autophagy in liver regeneration remain unclear. This study investigated the role of the essential autophagy gene encoding autophagy-related 16-like 1 (ATG16L1), which regulates the macrophage phenotype in liver regeneration. Methods We generated FloxP-Atg16l1 (Atg16l1 FL/FL ), Lyz2-Cre Atg16l1 knockout (KO) (Atg16l1 M-KO ), and myeloid-specific Atg16l1-overexpression-knock-in (Atg16l1 OE ) mice. These mice were subjected to 70% partial hepatectomy to demonstrate the role of ATG16L1 in macrophages during liver regeneration. Results ATG16L1 expression was significantly upregulated in macrophages during the early stages of liver regeneration. ATG16L1 deletion in macrophages substantially delayed liver regeneration in mice and caused a marked imbalance in Ly6Chi and Ly6Clo macrophage proportions in the liver. RNA-sequencing analysis revealed that, compared with macrophages isolated from Atg16l1 FL/FL mice, those from Atg16l1 M-KO mice exhibited significant downregulation of genes associated with oxidative phosphorylation and upregulation of proinflammatory gene expression. Mechanistically, ATG16L1 loss impaired mitophagy in macrophages, leading to the accumulation of mitochondrial damage and a metabolic shift that promoted proinflammatory macrophage polarization. ATG16L1 deficiency not only promoted macrophage mitochondrial (mt)DNA release and cyclic GMP-AMP synthase-stimulator of interferon genes (STING) activation, but also suppressed STING degradation. Sustained STING hyperactivation and subsequent increased release of downstream interferons further contributed to the inhibition of liver regeneration. Notably, pharmacological activation or genetic overexpression of ATG16L1 significantly enhanced liver regeneration in mice. Conclusions ATG16L1 has a pivotal role in liver regeneration by affecting the phenotype and function of macrophages. Thus, targeting ATG16L1 in macrophages could present a novel strategy for promoting liver regeneration. Impact and implications The autophagy-related gene ATG16L1 mediates mitophagy, facilitating the clearance of damaged mitochondria in macrophages following partial hepatectomy and maintaining a reparative macrophage phenotype. ATG16L1 deficiency triggers excessive STING activation and inhibits its degradation, thereby suppressing liver regeneration. Thus, targeting ATG16L1 in macrophages could represent a novel strategy to promote liver regeneration.
Collapse
Affiliation(s)
- Xinyu Zhan
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Yan Bai
- Department of Anesthesiology, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, 210029, China
| | - Qing Zhu
- Department of Anesthesiology, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, 210029, China
| | - Yiyun Gao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Fan Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Qingfa Bu
- Department of General Surgery, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zeyu Zhu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Zhuqing Rao
- Department of Anesthesiology, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, 210029, China
| | - Haoming Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| |
Collapse
|
17
|
Cui Y, Ma X, Wei J, Chen C, Shakir N, Guirram H, Dai Z, Anderson T, Ferguson D, Qiu S. MET receptor tyrosine kinase promotes the generation of functional synapses in adult cortical circuits. Neural Regen Res 2025; 20:1431-1444. [PMID: 39075910 PMCID: PMC11624886 DOI: 10.4103/nrr.nrr-d-23-01471] [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: 08/31/2023] [Revised: 01/02/2024] [Accepted: 04/20/2024] [Indexed: 07/31/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202505000-00026/figure1/v/2024-07-28T173839Z/r/image-tiff Loss of synapse and functional connectivity in brain circuits is associated with aging and neurodegeneration, however, few molecular mechanisms are known to intrinsically promote synaptogenesis or enhance synapse function. We have previously shown that MET receptor tyrosine kinase in the developing cortical circuits promotes dendritic growth and dendritic spine morphogenesis. To investigate whether enhancing MET in adult cortex has synapse regenerating potential, we created a knockin mouse line, in which the human MET gene expression and signaling can be turned on in adult (10-12 months) cortical neurons through doxycycline-containing chow. We found that similar to the developing brain, turning on MET signaling in the adult cortex activates small GTPases and increases spine density in prefrontal projection neurons. These findings are further corroborated by increased synaptic activity and transient generation of immature silent synapses. Prolonged MET signaling resulted in an increased α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid/N-methyl-D-aspartate (AMPA/NMDA) receptor current ratio, indicative of enhanced synaptic function and connectivity. Our data reveal that enhancing MET signaling could be an interventional approach to promote synaptogenesis and preserve functional connectivity in the adult brain. These findings may have implications for regenerative therapy in aging and neurodegeneration conditions.
Collapse
Affiliation(s)
- Yuehua Cui
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Xiaokuang Ma
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Jing Wei
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Chang Chen
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Neha Shakir
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Hitesch Guirram
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Zhiyu Dai
- Department of Medicine, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Trent Anderson
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Deveroux Ferguson
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Shenfeng Qiu
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| |
Collapse
|
18
|
Fromme M, Klebingat F, Ellis P, Strnad P. Alpha-1 antitrypsin deficiency-associated liver disease: From understudied disorder to the poster child of genetic medicine. Hepatol Commun 2025; 9:e0699. [PMID: 40227077 PMCID: PMC11999460 DOI: 10.1097/hc9.0000000000000699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/07/2025] [Indexed: 04/15/2025] Open
Abstract
Alpha-1 antitrypsin deficiency (AATD) constitutes an inborn disorder arising due to mutations in alpha-1 antitrypsin (AAT), a secreted protease inhibitor produced primarily in hepatocytes. It leads to diminished serum AAT levels, and this loss-of-function predisposes to chronic obstructive pulmonary disease and lung emphysema. The characteristic Pi*Z mutation results in hepatic Z-AAT accumulation. In its homozygous form (Pi*ZZ genotype), it is responsible for the majority of severe AATD cases and can cause both pediatric and adult liver disease, while the heterozygous form (Pi*MZ) is considered a disease modifier that becomes apparent primarily in the presence of other comorbidities or risk factors. In the current review, we collate conditions associated with AATD, introduce typical AAT variants, and discuss our understanding of disease pathogenesis. We present both cross-sectional and longitudinal data informing about the natural disease history and noninvasive tools that can be used for disease stratification as well as a basis for disease monitoring. Given that AATD-associated liver disease is highly heterogeneous, we discuss the risk factors affecting disease progression. While the loss-of-function lung disease is treated by weekly intravenous administration of purified AAT, recombinant modified AAT and oral protease inhibitors are currently in clinical trials. Among the liver candidates, small interfering RNA fazirsiran efficiently suppresses AAT production and is currently in phase 3 clinical trial, while several other genetic approaches, such as RNA editing, are at earlier stages. In summary, AATD represents a systemic disorder increasingly seen in the hepatologic routine and requiring thorough interdisciplinary care, since the currently ongoing clinical trials often address only one of the organs it affects.
Collapse
Affiliation(s)
- Malin Fromme
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN RARE LIVER), Aachen, Germany
| | - Fabienne Klebingat
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN RARE LIVER), Aachen, Germany
| | - Paul Ellis
- School of Health Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Pavel Strnad
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN RARE LIVER), Aachen, Germany
| |
Collapse
|
19
|
Vosough M, Shokouhian B, Sharbaf MA, Solhi R, Heidari Z, Seydi H, Hassan M, Devaraj E, Najimi M. Role of mitogens in normal and pathological liver regeneration. Hepatol Commun 2025; 9:e0692. [PMID: 40304568 PMCID: PMC12045551 DOI: 10.1097/hc9.0000000000000692] [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: 12/16/2024] [Accepted: 01/31/2025] [Indexed: 05/02/2025] Open
Abstract
The liver has a unique ability to regenerate to meet the body's metabolic needs, even following acute or chronic injuries. The cellular and molecular mechanisms underlying normal liver regeneration have been well investigated to improve organ transplantation outcomes. Once liver regeneration is impaired, pathological regeneration occurs, and the underlying cellular and molecular mechanisms require further investigations. Nevertheless, a plethora of cytokines and growth factor-mediated pathways have been reported to modulate physiological and pathological liver regeneration. Regenerative mitogens play an essential role in hepatocyte proliferation. Accelerator mitogens in synergism with regenerative ones promote liver regeneration following hepatectomy. Finally, terminator mitogens restore the proliferating status of hepatocytes to a differentiated and quiescent state upon completion of regeneration. Chronic loss of hepatocytes, which can manifest in chronic liver disorders of any etiology, often has undesired structural consequences, including fibrosis, cirrhosis, and liver neoplasia due to the unregulated proliferation of remaining hepatocytes. In fact, any impairment in the physiological function of the terminator mitogens results in the progression of pathological liver regeneration. In the current review, we intend to highlight the updated cellular and molecular mechanisms involved in liver regeneration and discuss the impairments in central regulating mechanisms responsible for pathological liver regeneration.
Collapse
Affiliation(s)
- Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bahare Shokouhian
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Amin Sharbaf
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Roya Solhi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Heidari
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Homeyra Seydi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ezhilarasan Devaraj
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| |
Collapse
|
20
|
Liu Q, Yu J, Chen L, Han J, Cai X, Hu S, Chu X, Zhang W, Wang Z. A deep-red hemicyanine fluorescent probe for imaging butyrylcholinesterase in living cells and in mice with APAP-induced liver injury. Talanta 2025; 286:127478. [PMID: 39765081 DOI: 10.1016/j.talanta.2024.127478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/10/2024] [Accepted: 12/26/2024] [Indexed: 03/03/2025]
Abstract
As a serine hydrolase synthesized by the liver, butyrylcholinesterase (BChE) is an important biomarker in the clinical diagnosis of liver diseases. To track BChE activity in drug-induced liver injury, we designed a deep-red BChE-activatable fluorescent probe (CYL-BChE) with hemi-cyanine structure by using a cyclopropyl carbonyl group as a specific recognition moiety. Its near-infrared absorption wavelength (665 nm) and emission wavelength (762 nm) provide excellent tissue penetration capabilities, making it suitable for biological imaging. Additionally, CYL-BChE has a large Stokes shift (97 nm) and a low detection limit (0.96 U/L), effectively distinguishing BChE from the interfering substance acetylcholinesterase (AChE). Besides of detection of endogenous BChE in live cells, the resulting probe has been successfully used to detect BChE expression levels in an acetaminophen-induced (APAP-induced) drug liver injury mouse model. Therefore, this probe is expected to be a valuable biological tool in the detection of BChE activity.
Collapse
Affiliation(s)
- Qi Liu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Jiaying Yu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Lucheng Chen
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Jiaxing Han
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Xiangyu Cai
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Shaojun Hu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Xianfeng Chu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Weijie Zhang
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Zhifei Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
21
|
El Raziky M, Rashwan M, Tamim HH, Afify S, Lithy RM. Role of IL-6, TNF-α and VCAM-1 as predictors of renal impairment in patients with spontaneous bacterial peritonitis. Trans R Soc Trop Med Hyg 2025; 119:524-533. [PMID: 39758003 DOI: 10.1093/trstmh/trae139] [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: 07/07/2024] [Revised: 10/17/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Multiple mechanisms may contribute to the occurrence of renal impairment (RI) in patients with spontaneous bacterial peritonitis (SBP). One such mechanism is systemic inflammatory response syndrome, which involves the release of pro-inflammatory cytokines (tumour necrosis factor [TNF]-α, interleukin [IL]-6 and vascular cell adhesion molecule [VCAM]-1). The goal of this research was to evaluate the role of IL-6, TNF-α and VCAM-1 as potential predictors of RI and mortality in cirrhotic patients with SBP. METHODS This study included 90 cirrhotic patients with SBP, divided into two equal groups: group A was patients without RI and group B was patients with RI. Based on mortality outcomes, the patients were further categorized into group 1 (recovery, n=70) and group 2 (death, n=20). TNF-α, IL-6 and VCAM-1 serum levels were measured using enzyme-linked immunosorbent assay. RESULTS RI occurred in 50% (45/90) of the study population. Among the 90 patients, 10 (11.1%) had elevated IL-6 levels, 8 (8.9%) had elevated TNF-α levels and 6 (6.7%) had elevated VCAM-1 levels. There were no significant variations in cytokine levels between groups A and B. With an area under the curve of 0.5, the three cytokines showed comparable sensitivity and specificity as predictors of RI. CONCLUSIONS The use of TNF-α, IL-6 and VCAM-1 as predictive markers for RI and mortality in SBP patients is not recommended, as these biomarkers demonstrated limited diagnostic value.
Collapse
Affiliation(s)
- Maissa El Raziky
- Endemic Medicine and Hepato-Gastroenterology Department, Faculty of Medicine, Cairo University, Cairo 3753450, Egypt
| | - Mennatallah Rashwan
- Endemic Medicine and Hepato-Gastroenterology Department, Faculty of Medicine, Cairo University, Cairo 3753450, Egypt
| | - Hend Hamed Tamim
- Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo 3753450, Egypt
| | - Shimaa Afify
- Department of Gastroenterology, National Hepatology and Tropical Medicine Research Institute, Cairo 3753450, Egypt
| | - Rania M Lithy
- Endemic Medicine and Hepato-Gastroenterology Department, Faculty of Medicine, Cairo University, Cairo 3753450, Egypt
| |
Collapse
|
22
|
Zhuang X, Chen X, Cao L, Wang B, Wang Z, Li S, Li H, Li C, Yang N. The class A scavenger receptor member 3 (SCARA3) regulates cell apoptosis through X-linked apoptosis inhibitory protein (XIAP) in turbot (Scophthalmus maximus L.). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 166:105370. [PMID: 40194751 DOI: 10.1016/j.dci.2025.105370] [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: 02/04/2025] [Revised: 03/29/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
Class A scavenger receptor 3 (SCARA3), a macrophage scavenger receptor-like protein, plays important roles in inhibiting cell proliferation, migration, and invasion. In the present study, a SCARA3 gene of turbot (SmSCARA3) (Gene ID: 118289953) with an 1815 bp ORF encoding 604 amino acids was identified. Phylogenetic analysis revealed that SmSCARA3 showed the closest relationship to that counterpart of olive flounder (Paralichthys olivaceus). The synteny analysis demonstrated conserved syntenic patterns across selected vertebrates. In addition, SmSCARA3 was ubiquitously expressed in all the examined tissues, with the highest expression level in intestine and the lowest expression level in the brain. SmSCARA3 exhibited different expression patterns in mucosal tissues (intestine, gill, skin) after two bacterial infections. Subsequently, recombinant SmSCARA3 protein (rSmSCARA3) revealed the strong binding affinity to LPS and responded primarily to LPS stimulation in intestinal cells of turbot. Additionally, the interference and overexpression experiments indicated that SmSCARA3 was associated with apoptosis related genes, such as Caspase1, Caspase3 and Caspase3a, and it could activate Caspase3 in HEPG2 cells. Moreover, flow cytometry revealed the apoptosis of SmSCARA3 overexpression group increased by 10.03%, which was consistent with the effect of SmSCARA3 on proliferation inhibition in intestinal cells of turbot. The cell apoptosis levels in the SmSCARA3-Flag and XIAP-HA experimental group were significantly lower than that in the control group (51.17% vs 72.72%). Finally, the Co-IP assay showed that SmSCARA3 could directly interact with XIAP. In conclusion, our results indicated that SmSCARA3 could activate Caspase3 and modulate apoptosis through XIAP , highlighting its potential roles as a therapeutic target for fish diseases.
Collapse
Affiliation(s)
- Xinghua Zhuang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Xuan Chen
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Lili Cao
- Shandong Freshwater Fisheries Research Institute, Jinan, 250013, China
| | - Beibei Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Zhongyi Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Suwan Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Honghong Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chao Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| | - Ning Yang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
23
|
Liao C, Zhang Y, Yang J, Wang S, Li Z, Chen S, Xie Y, Xu L, Peng S, Zeng X, Kuang M, Xiang B, Sun K, Zhao X. Single-Cell Transcriptomic Analysis Reveals an Aggressive Basal-Like Tumor Cell Subpopulation Associated With Poor Prognosis in Intrahepatic Cholangiocarcinoma. J Gastroenterol Hepatol 2025; 40:1263-1273. [PMID: 39993788 DOI: 10.1111/jgh.16915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/03/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND AND AIM Intrahepatic cholangiocarcinoma (ICC) is the second most common primary liver cancer whose incidence is increasing globally. However, the high tumor heterogeneity of ICC restricts the efficacy of available systematic therapies. We aim to dissect the tumor heterogeneity of ICC utilizing high-resolution single-cell RNA sequencing to identify novel therapeutic targets. METHODS We performed single-cell RNA sequencing (scRNA-seq) of 26 tumor samples from 23 ICC patients and spatial transcriptomic sequencing of six tumor sections from six ICC patients. Bulk RNA-seq data from two public datasets were used for validation. Additionally, immunohistochemical staining and multiplex immunofluorescence staining were conducted to validate the infiltration and distribution of cells in the tumor microenvironment. RESULTS We discovered that malignant cells in ICC samples exhibited a remarkably high degree of tumor heterogeneity. We identified a basal-like tumor cell subpopulation characterized by the expression of basal epithelial related genes including KRT5, KRT6A, and KRT17. The basal-like tumor subpopulation was characterized by activation of MET signaling and extracellular matrix organization associated with tumor invasion and correlated with poor prognosis. Cell-cell communication analysis further showed significant HGF-MET interaction between inflammatory cancer-associated fibroblasts (iCAFs) and basal-like tumor cells. We found that iCAFs were the major source of HGF in tumor environment and contributed to the basal-like phenotype formation of tumor cells by HGF-MET axis. CONCLUSIONS We identified an aggressive basal-like tumor cell subpopulation, which correlated with poor prognosis in ICC. The MET pathway contributes to the aggressiveness of basal-like tumor cells and serves as a novel therapeutic target for ICC.
Collapse
Affiliation(s)
- Changyi Liao
- Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuting Zhang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jing Yang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuo Wang
- Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhijuan Li
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuling Chen
- Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yubin Xie
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lixia Xu
- Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Sui Peng
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Clinical Trials Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xuezhen Zeng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ming Kuang
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Bangde Xiang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guang xi, China
| | - Kaiyu Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao Zhao
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
24
|
De Rudder M, Manco R, Coubeau L, Fontaine A, Bertrand C, Leclercq IA, Dili A. Vascular damage and excessive proliferation compromise liver function after extended hepatectomy in mice. Hepatology 2025; 81:1468-1484. [PMID: 38661628 DOI: 10.1097/hep.0000000000000900] [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: 10/12/2023] [Accepted: 03/27/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND AND AIMS Surgical resection remains the gold standard for liver tumor treatment, yet the emergence of postoperative liver failure, known as the small-for-size syndrome (SFSS), poses a significant challenge. The activation of hypoxia sensors in an SFSS liver remnant initiated early angiogenesis, improving the vascular architecture, safeguarding against liver failure, and reducing mortality. The study aimed to elucidate vascular remodeling mechanisms in SFSS and their impact on hepatocyte function and subsequent liver failure. APPROACH AND RESULTS Mice underwent extended partial hepatectomy to induce SFSS, with a subset exposed to hypoxia immediately after surgery. Hypoxia bolstered posthepatectomy survival rates. The early proliferation of liver sinusoidal cells, coupled with recruitment of putative endothelial progenitor cells, increased vascular density, improved lobular perfusion, and limited hemorrhagic events in the regenerating liver under hypoxia. Administration of granulocyte colony-stimulating factor in hepatectomized mice mimicked the effects of hypoxia on vascular remodeling and endothelial progenitor cell recruitment but failed to rescue survival. Compared to normoxia, hypoxia favored hepatocyte function over proliferation, promoting functional preservation in the regenerating remnant. Injection of Adeno-associated virus serotype 8-thyroxine-binding globulin-hepatocyte nuclear factor 4 alpha virus for hepatocyte-specific overexpression of hepatocyte nuclear factor 4 alpha, the master regulator of hepatocyte function, enforced functionality in proliferating hepatocytes but did not rescue survival. The combination of hepatocyte nuclear factor 4 alpha overexpression and granulocyte colony-stimulating factor treatment rescued survival after SFSS-setting hepatectomy. CONCLUSIONS In summary, SFSS arises from an imbalance and desynchronized interplay between functional regeneration and vascular restructuring. To improve survival following SFSS hepatectomy, it is essential to adopt a 2-pronged strategy aimed at preserving the function of proliferating parenchymal cells and simultaneously attenuating vascular damage.
Collapse
Affiliation(s)
- Maxime De Rudder
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Rita Manco
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Laurent Coubeau
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Surgery, University Clinics of St Luc, UCLouvain, Brussels, Belgium
| | - Alix Fontaine
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Claude Bertrand
- Department of Surgery, University Hospital of UCLouvain-Namur, Site of Godinne, Yvoir, Belgium
| | - Isabelle A Leclercq
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Alexandra Dili
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Surgery, University Hospital of UCLouvain-Namur, Site of Godinne, Yvoir, Belgium
| |
Collapse
|
25
|
Ortuño-Costela MC, Pinzani M, Vallier L. Cell therapy for liver disorders: past, present and future. Nat Rev Gastroenterol Hepatol 2025; 22:329-342. [PMID: 40102584 DOI: 10.1038/s41575-025-01050-2] [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] [Accepted: 02/11/2025] [Indexed: 03/20/2025]
Abstract
The liver fulfils a plethora of vital functions and, due to their importance, liver dysfunction has life-threatening consequences. Liver disorders currently account for more than two million deaths annually worldwide and can be classified broadly into three groups, considering their onset and aetiology, as acute liver diseases, inherited metabolic disorders and chronic liver diseases. In the most advanced and severe forms leading to liver failure, liver transplantation is the only treatment available, which has many associated drawbacks, including a shortage of organ donors. Cell therapy via fully mature cell transplantation is an advantageous alternative that may be able to restore a damaged organ's functionality or serve as a bridge until regeneration can occur. Pioneering work has shown that transplanting adult hepatocytes can support liver recovery. However, primary hepatocytes cannot be grown extensively in vitro as they rapidly lose their metabolic activity. Therefore, different cell sources are currently being tested as alternatives to primary cells. Human pluripotent stem cell-derived cells, chemically induced liver progenitors, or 'liver' organoids, hold great promise for developing new cell therapies for acute and chronic liver diseases. This Review focuses on the advantages and drawbacks of distinct cell sources and the relative strategies to address different therapeutic needs in distinct liver diseases.
Collapse
Affiliation(s)
- M Carmen Ortuño-Costela
- Berlin Institute of Health, BIH Centre for Regenerative Therapies, Charité-Universitätsmedizin, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Massimo Pinzani
- University College London Institute for Liver and Digestive Health, Division of Medicine, Royal Free Hospital, London, UK
- University of Pittsburgh Medical Center-Mediterranean Institute for Transplantation and Highly Specialized Therapies (UPMC-ISMETT), Palermo, Italy
| | - Ludovic Vallier
- Berlin Institute of Health, BIH Centre for Regenerative Therapies, Charité-Universitätsmedizin, Berlin, Germany.
- Max Planck Institute for Molecular Genetics, Berlin, Germany.
| |
Collapse
|
26
|
Zhang X, Li S, Hao L, Jia F, Yu F, Hu X. Influencing factors and mechanism of hepatocyte regeneration. J Transl Med 2025; 23:493. [PMID: 40307789 PMCID: PMC12042435 DOI: 10.1186/s12967-025-06278-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/20/2025] [Indexed: 05/02/2025] Open
Abstract
As a research hotspot in the field of regenerative medicine, hepatocyte regeneration has great potential in the treatment of liver diseases. This paper comprehensively summarizes the diverse sources of hepatocyte regeneration and its complex influencing factors, and deeply discusses the typical mechanism. According to the existing research, we observed that Wnt signaling pathway and Notch signaling pathway can play a synergistic role in the process of hepatocyte regeneration. So we further analyzed the crosstalk between Wnt and Notch signal pathway and the cross mechanism with TGF-β, YAP/TAZ pathway during regeneration. Despite the remarkable progress in the study of liver regeneration at the cellular and molecular levels, the comprehensive understanding of the fine regulation of influencing factors and the interaction between mechanisms still needs to be deepened. This paper aims to systematically analyze the interaction between influencing factors and classical mechanisms of hepatocyte regeneration by integrating multi-group data and advanced bioinformatics methods, so as to provide feasible ideas for the treatment of liver diseases and lay a solid theoretical foundation for the future development of regenerative medicine. It is believed that focusing on the rational development of innovative means such as inducing gene tendentiousness expression and anti-aging therapy, and in-depth analysis of the complex interactive network between hepatocyte regeneration mechanisms are expected to open up a new road for the development of more effective treatment strategies for liver diseases.
Collapse
Affiliation(s)
- Xiaoyi Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, No.39, Shierqiao Road, Jinniu District, Chengdu, Sichuan, China
- Clinical Medical College of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Shenghao Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, No.39, Shierqiao Road, Jinniu District, Chengdu, Sichuan, China
- Clinical Medical College of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Liyuan Hao
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, No.39, Shierqiao Road, Jinniu District, Chengdu, Sichuan, China
- Clinical Medical College of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Fukang Jia
- Henan University of Traditional Chinese, Zhengzhou, China
| | - Fei Yu
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, No.39, Shierqiao Road, Jinniu District, Chengdu, Sichuan, China
- Clinical Medical College of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyu Hu
- Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Department of Infectious Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, No.39, Shierqiao Road, Jinniu District, Chengdu, Sichuan, China.
| |
Collapse
|
27
|
P S, P R R, K P. Feature selection for classification based on machine learning algorithms for prostate cancer. Biomed Phys Eng Express 2025; 11:035023. [PMID: 40262626 DOI: 10.1088/2057-1976/adcf2b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 04/22/2025] [Indexed: 04/24/2025]
Abstract
Microarray technology has transformed the biotechnological research to next level in the recent years. It provides the expression levels of various genes involved in a particular disease. Prostate cancer disease turned into life threatening cancer. The genes causing this disease are identified through the classification methods. These gene expression data have problems like high dimensional with low sample size which imposes active challenges in the existing classification algorithms. Feature selection techniques are applied in order to address the dimensionality issues. . This paper aims in analyzing the feature selection methods for classification of gene expression data of Prostate and identify the significant genes that have a major influence on the disease. The three different feature selection methods such as Filters, wrappers and embedded selectors are applied before the classification process for selecting the top ranked genes. Then, the extracted top ranked genes are applied on the classification algorithms such as SVM, k-NN, Random Forest and Artificial Neural Network. After the inclusion of feature selection technique, the classification accuracy is significantly boosted even with less number of genes. Random Forest classification algorithm outperforms other classification methods. The significant genes that has the major influence in prostate cancer disease are identified such as KLK3, GFI1, CXCR2 and TNFRSF10C.
Collapse
Affiliation(s)
- Swathypriyadharsini P
- Department of Computer Science and Engineering, Bannari Amman Institute of Technology, Sathyamangalam, Erode, Tamil Nadu, India
| | - Rupashini P R
- Department of Artificial Intelligence and Data Science, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, India
| | - Premalatha K
- Department of Computer Science and Engineering, Bannari Amman Institute of Technology, Sathyamangalam, Erode, Tamil Nadu, India
| |
Collapse
|
28
|
Yang C, Gao Z, Tang R, Zhou L, Zhou P, Shi W, Ren D, Chen H, Zhang Z, Xie X, Lin J, Ye Y, Feng Z, Feng X, He Y, Zhi Y, Liu D, Zhang X, Jiang L. POU6F2 promotes liver metastasis of gastric adenocarcinoma by dual mechanism of transcriptional upregulation of SNAI1 and IGF2/PI3K/AKT signaling-induced conversion of hepatic stellate cells into cancer-associated fibroblasts. Br J Cancer 2025:10.1038/s41416-025-03017-1. [PMID: 40301686 DOI: 10.1038/s41416-025-03017-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 03/10/2025] [Accepted: 04/02/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Activation of cancer-associated fibroblasts (CAFs) plays an important role in tumor metastasis. The purpose of this study is to investigate the role of POU6F2 in conversion of hepatic stellate cells (HSCs) into CAFs in liver metastasis of gastric adenocarcinoma (GAC). METHODS POU6F2 expression was examined by real-time PCR, Western blot and immunohistochemical staining. The functional roles of POU6F2 in GAC liver metastasis were investigated both cellular experiments in vitro and in vivo using a mouse model of subcutaneous splenic injection. ChIP and ELISA assays were used to explore the underlying molecular mechanism of POU6F2 in liver metastasis of GAC. RESULTS Here we reported that POU6F2 was upregulated in GAC tissue with liver metastasis, which predicted poor early liver metastasis. Upregulating POU6F2 promoted EMT, invasion and migration of GAC cells in vitro, and the liver metastasis of GAC cells in vivo. Mechanic investigation further revealed that upregulating POU6F2 promoted the invasion and metastasis of GAC by transcriptional upregulation of EMT-inducer SNAI1, and promoting the conversion of HSCs into CAFs dependent on transcriptional upregulation of IGF2-induced activation of PI3K/AKT signaling. CONCLUSION Our findings uncover a novel dual mechanism by which POU6F2 promotes liver metastasis of GAC.
Collapse
Affiliation(s)
- Chunxiao Yang
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Zhiqing Gao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
- Department of Molecular Medicine, School of Basic Medicine, Southern Medical University, Guangzhou, China
| | - Ruiming Tang
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Lihuan Zhou
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Ping Zhou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Wangpan Shi
- Department of Pathology, University of California San Diego Health System, San Diego, CA, USA
| | - Dong Ren
- Departments of Pathology, University of California Irvine Medical Center, Orange, CA, USA
| | - Han Chen
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Zhuojun Zhang
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Xiaoyi Xie
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Jiaqian Lin
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Yingming Ye
- Department of Pathogen Biology and Immunology, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhengfu Feng
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Xiaoli Feng
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Yaoming He
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Jiangmen, China
| | - Yaofeng Zhi
- Clinical Experimental Center, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Clinical Medical College of Guangdong Medical University, Jiangmen Central Hospital, Jiangmen, China
| | - Dongmei Liu
- Clinical Experimental Center, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Clinical Medical College of Guangdong Medical University, Jiangmen Central Hospital, Jiangmen, China
| | - Xin Zhang
- Clinical Experimental Center, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Clinical Medical College of Guangdong Medical University, Jiangmen Central Hospital, Jiangmen, China.
| | - Lili Jiang
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China.
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
29
|
Subramaniam S, Jeet V, Gunter JH, Janjua Khan T, Feng Y, Clements JA, Srinivasan S, Popat A, Batra J. Lactoferrin-encapsulated dichloroacetophenone (DAP) nanoparticles enhance drug delivery and anti-tumor efficacy in prostate cancer. Cancer Lett 2025; 616:217522. [PMID: 39924080 DOI: 10.1016/j.canlet.2025.217522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
Pyruvate Dehydrogenase Kinase 1 (PDK1) regulates glycolysis and oxidative phosphorylation pathways and is linked to prostate cancer metastasis and poor prognosis. The therapeutic application of 2,2-dichloroacetophenone (DAP), a PDK1 inhibitor, remains underexplored in prostate cancer. In this study we demonstrated that DAP exhibited a superior ability to inhibit prostate cancer cell proliferation, migration and colony formation at a lower concentration (20 μM) compared to a previously established inhibitor, dichloroacetate (DCA), which required concentrations of 30 mM or higher. However, poor aqueous solubility and lower stability of DAP limits its therapeutic application. Nano formulation of DAP with natural lactoferrin enhanced its dispersion and stability by increasing polydispersity index and intensity, and reduced zeta potential values upon conjugation that overcame the solubility limitations of DAP. The lactoferrin-DAP nanoparticles exhibited enhanced therapeutic efficacy by precisely targeting prostate cancer cells that express high lactoferrin receptors and high anti-tumor activity in vitro (at 1 μM) and in mouse prostate tumor xenografts (20 mg/kg). Mechanistically, these nanoparticles induce apoptosis in cancer cells by inducing caspase3/7 activity and disrupting the glycolytic and oxidative phosphorylation pathways. Moreover, lactoferrin-conjugated DAP nanoparticles suppressed the viability of docetaxel-resistant cells exhibiting a higher inhibitory efficacy compared to free DAP and DCA. Targeting PDK1 through lactoferrin-conjugated DAP nanoparticles represents a potent targeted therapeutic strategy for disrupting prostate tumor metabolism and offers promising implications for overcoming drug resistance.
Collapse
Affiliation(s)
- Sugarniya Subramaniam
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland (QLD), Australia; Translational Research Institute, Woolloongabba, QLD, Australia
| | - Varinder Jeet
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland (QLD), Australia
| | - Jennifer H Gunter
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland (QLD), Australia; Translational Research Institute, Woolloongabba, QLD, Australia; The Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD, 4059, Australia
| | | | - Yuran Feng
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| | - Judith A Clements
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland (QLD), Australia; Translational Research Institute, Woolloongabba, QLD, Australia
| | - Srilakshmi Srinivasan
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland (QLD), Australia; Translational Research Institute, Woolloongabba, QLD, Australia; The Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD, 4059, Australia
| | - Amirali Popat
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia; Mater Research Institute, Woolloongabba, QLD, Australia; Department of Functional Materials and Catalysis, Faculty of Chemistry, University of Vienna, Währinger Straße 42, 1090, Vienna, Austria
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland (QLD), Australia; Translational Research Institute, Woolloongabba, QLD, Australia; The Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD, 4059, Australia.
| |
Collapse
|
30
|
Hammond NL, Murtuza Baker S, Georgaka S, Al-Anbaki A, Jokl E, Simpson K, Sanchez-Alvarez R, Athwal VS, Purssell H, Siriwardena AK, Spiers HVM, Dixon MJ, Bere LD, Jones AP, Haley MJ, Couper KN, Bobola N, Sharrocks AD, Hanley NA, Rattray M, Piper Hanley K. Spatial gene regulatory networks driving cell state transitions during human liver disease. EMBO Mol Med 2025:10.1038/s44321-025-00230-6. [PMID: 40281306 DOI: 10.1038/s44321-025-00230-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Liver fibrosis is a major cause of death worldwide. As a progressive step in chronic liver disease, fibrosis is almost always diagnosed too late with limited treatment options. Here, we uncover the spatial transcriptional landscape driving human liver fibrosis using single nuclei RNA and Assay for Transposase-Accessible Chromatin (ATAC) sequencing to deconvolute multi-cell spatial transcriptomic profiling in human liver cirrhosis. Through multi-modal data integration, we define molecular signatures driving cell state transitions in liver disease and define an impaired cellular response and directional trajectory between hepatocytes and cholangiocytes associated with disease remodelling. We identify pro-fibrogenic signatures in non-parenchymal cell subpopulations co-localised within the fibrotic niche and localise transitional cell states at the scar interface. This combined approach provides a spatial atlas of gene regulation and defines molecular signatures associated with liver disease for targeted therapeutics or as early diagnostic markers of progressive liver disease.
Collapse
Affiliation(s)
- Nigel L Hammond
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Syed Murtuza Baker
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Sokratia Georgaka
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Ali Al-Anbaki
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Elliot Jokl
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Kara Simpson
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Rosa Sanchez-Alvarez
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Varinder S Athwal
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
- Manchester University NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Huw Purssell
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
- Manchester University NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Ajith K Siriwardena
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
- Manchester University NHS Foundation Trust, Oxford Road, Manchester, UK
| | | | - Mike J Dixon
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Leoma D Bere
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Adam P Jones
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Michael J Haley
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Kevin N Couper
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Nicoletta Bobola
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Andrew D Sharrocks
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Neil A Hanley
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
- College of Medical & Dental Sciences, University of Birmingham, Birmingham, UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2GW, UK
| | - Magnus Rattray
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Karen Piper Hanley
- Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK.
| |
Collapse
|
31
|
Lian J, An Y, Wei W, Lu Y, Zhang X, Sun G, Guo H, Xu L, Chen X, Hu H. Transcriptional landscape and chromatin accessibility reveal key regulators for liver regenerative initiation and organoid formation. Cell Rep 2025; 44:115633. [PMID: 40286271 DOI: 10.1016/j.celrep.2025.115633] [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: 04/23/2024] [Revised: 03/19/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Liver regeneration is a well-organized and phase-restricted process that involves chromatin remodeling and transcriptional alterations. However, the specific transcription factors (TFs) that act as key "switches" to initiate hepatocyte regeneration and organoid formation remain unclear. Comprehensive integration of RNA sequencing and ATAC sequencing reveals that ATF3 representing "Initiation_on" TF and ONECUT2 representing "Initiation_off" TF transiently modulate the occupancy of target promoters to license liver cells for regeneration. Knockdown of Atf3 or overexpression of Onecut2 not only reduces organoid formation but also delays tissue-damage repair after PHx or CCl4 treatment. Mechanistically, we demonstrate that ATF3 binds to the promoter of Slc7a5 to activate mTOR signals while the Hmgcs1 promoter loses ONECUT2 binding to facilitate regenerative initiation. The results identify the mechanism for initiating regeneration and reveal the remodeling of transcriptional landscapes and chromatin accessibility, thereby providing potential therapeutic targets for liver diseases with regenerative defects.
Collapse
Affiliation(s)
- Jiabei Lian
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yachun An
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Wenjing Wei
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yao Lu
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Xiyu Zhang
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Gongping Sun
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Histology and Embryology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Haiyang Guo
- Department of Clinical Laboratory, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Longjin Xu
- Shandong Center for Disease Control and Prevention, Jinan, Shandong 250014, China
| | - Xuena Chen
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Huili Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
32
|
Modares NF, Hendrikse LD, Smith LK, Paul MS, Haight J, Luo P, Liu S, Fortin J, Tong FK, Wakeham AC, Jafari SM, Zheng C, Buckland M, Flick R, Silvester J, Berger T, Ketela T, Helke S, Foffi E, Niavarani R, Mcwilliam R, Saunders ME, Colonna I, David BA, Rastogi T, Lee WY, Kubes P, Mak TW. B cell-derived acetylcholine promotes liver regeneration by regulating Kupffer cell and hepatic CD8 + T cell function. Immunity 2025:S1074-7613(25)00165-7. [PMID: 40286791 DOI: 10.1016/j.immuni.2025.04.002] [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: 07/09/2024] [Revised: 12/20/2024] [Accepted: 04/02/2025] [Indexed: 04/29/2025]
Abstract
Liver regeneration (LR) is essential for recovery from acute trauma, cancer surgery, or transplantation. Neurotransmitters such as acetylcholine (ACh) play a role in LR by stimulating immune cells and augmenting hepatocyte proliferation, but the source of this ACh remains unclear. Here, we demonstrated that B cells expressing choline acetyltransferase (ChAT), which synthesizes ACh, were required for LR. Mice lacking ChAT+ B cells subjected to partial hepatectomy (PHX) displayed greater mortality due to failed LR. Kupffer cells and hepatic CD8+ T cells expressed the α7 nicotinic ACh receptor (nAChR), and LR was disrupted in mice lacking α7 nAChR. Mechanistically, B cell-derived ACh signaled through α7 nAChR to positively regulate the function of regenerative Kupffer cells and to control the activation of hepatic CD8+ T cells to curtail harmful interferon-gamma (IFNγ) production. Our work offers insights into LR mechanisms that may point to therapies for liver damage.
Collapse
Affiliation(s)
| | - Liam D Hendrikse
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Logan K Smith
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Michael St Paul
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jillian Haight
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ping Luo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Shaofeng Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jerome Fortin
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Frances K Tong
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Andrew C Wakeham
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Chunxing Zheng
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China
| | - Mackenzie Buckland
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Robert Flick
- Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Jennifer Silvester
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Thorsten Berger
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Troy Ketela
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Simone Helke
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Erica Foffi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Raheleh Niavarani
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ryan Mcwilliam
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Mary E Saunders
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Isabelle Colonna
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Bruna Araujo David
- Calvin Phoebe & Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Physiology and Pharmacology Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Tashi Rastogi
- Calvin Phoebe & Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Physiology and Pharmacology Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Woo-Yong Lee
- Department of Biomedical and Molecular Science, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Paul Kubes
- Calvin Phoebe & Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Physiology and Pharmacology Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Biomedical and Molecular Science, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Tak W Mak
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China; Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China; Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
33
|
Zhao S, Bai B, Zhu B, Cui Y, Deng W, Xie Z, Wang S, Wang X, Mao Y, Lu Y, Huang Y, You T, Sun W, Shen X, Lu X. Hyaluronic acid-modified doxorubicin-covalent organic framework nanoparticles triggered pyroptosis in combinations with immune checkpoint blockade for the treatment of breast cancer. Int J Biol Macromol 2025; 310:143265. [PMID: 40274165 DOI: 10.1016/j.ijbiomac.2025.143265] [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: 02/19/2025] [Revised: 04/03/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive malignancy, and the current treatment strategies have poor efficacy. Pyroptosis, a type of immunogenic cell death, significantly enhances antitumor immunity by triggering the release of numerous intracellular components. Here, we prepared a covalent-organic framework (COF) loaded with doxorubicin (DOX) and coated it with hyaluronic acid (HA) as a therapeutic delivery system (HA@DOX-COF) for the treatment of TNBC. Mechanistically, upon cellular uptake, HA@DOX-COF activated pyroptosis through the caspase-3/GSDME pathway. Moreover, HA@DOX-COF induced a pyroptosis-induced antitumor immune response and further augmented the efficacy of immune checkpoint blockade (ICB). Our results demonstrated that the combination of HA@DOX-COF and an anti-PD-1 antibody markedly inhibited tumor progression in a TNBC murine model. Overall, our work offers a potential approach for pyroptosis-induced immunotherapy for TNBC, which may increase the efficacy of ICB-based immunotherapy.
Collapse
Affiliation(s)
- Shengsheng Zhao
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Binglong Bai
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Bingzi Zhu
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yuekai Cui
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Wenhai Deng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Zuoliang Xie
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Shuaibin Wang
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiang Wang
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yiwen Mao
- Department of Breast Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yongyong Lu
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yingpeng Huang
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Tao You
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Weijian Sun
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Xian Shen
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Xufeng Lu
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
34
|
Lee LR, Guillotin B, Rahni R, Hutchison C, Desvoyes B, Gutierrez C, Birnbaum KD. Glutathione accelerates the cell cycle and cellular reprogramming in plant regeneration. Dev Cell 2025; 60:1153-1167.e6. [PMID: 39755116 DOI: 10.1016/j.devcel.2024.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/01/2024] [Accepted: 12/10/2024] [Indexed: 01/06/2025]
Abstract
The plasticity of plant cells underlies their wide capacity to regenerate, with increasing evidence in plants and animals implicating cell-cycle dynamics in cellular reprogramming. To investigate the cell cycle during cellular reprogramming, we developed a comprehensive set of cell-cycle-phase markers in the Arabidopsis root. Using single-cell RNA sequencing profiles and live imaging during regeneration, we found that a subset of cells near an ablation injury dramatically increases division rate by truncating G1 phase. Cells in G1 undergo a transient nuclear peak of glutathione (GSH) prior to coordinated entry into S phase, followed by rapid divisions and cellular reprogramming. A symplastic block of the ground tissue impairs regeneration, which is rescued by exogenous GSH. We propose a model in which GSH from the outer tissues is released upon injury, licensing an exit from G1 near the wound to induce rapid cell division and reprogramming.
Collapse
Affiliation(s)
- Laura R Lee
- New York University, Center for Genomics and Systems Biology, Department of Biology, New York, NY 10003, USA
| | - Bruno Guillotin
- New York University, Center for Genomics and Systems Biology, Department of Biology, New York, NY 10003, USA
| | - Ramin Rahni
- New York University, Center for Genomics and Systems Biology, Department of Biology, New York, NY 10003, USA
| | - Chanel Hutchison
- New York University, Center for Genomics and Systems Biology, Department of Biology, New York, NY 10003, USA
| | | | | | - Kenneth D Birnbaum
- New York University, Center for Genomics and Systems Biology, Department of Biology, New York, NY 10003, USA.
| |
Collapse
|
35
|
Awasthi BW, Paulo JA, Burkhart DL, Smith IR, Collins RL, Harper JW, Gygi SP, Haigis KM. The network response to Egf is tissue-specific. iScience 2025; 28:112146. [PMID: 40171493 PMCID: PMC11960661 DOI: 10.1016/j.isci.2025.112146] [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: 04/22/2024] [Revised: 07/29/2024] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
Epidermal growth factor receptor (Egfr)-driven signaling regulates fundamental homeostatic processes. Dysregulated signaling via Egfr is implicated in numerous disease pathologies and distinct Egfr-associated disease etiologies are known to be tissue-specific. The molecular basis of this tissue-specificity remains poorly understood. Most studies of Egfr signaling to date have been performed in vitro or in tissue-specific mouse models of disease, which has limited insight into Egfr signaling patterns in healthy tissues. Here, we carried out integrated phosphoproteomic, proteomic, and transcriptomic analyses of signaling changes across various mouse tissues in response to short-term stimulation with the Egfr ligand Egf. We show how both baseline and Egf-stimulated signaling dynamics differ between tissues. Moreover, we propose how baseline phosphorylation and total protein levels may be associated with clinically relevant tissue-specific Egfr-associated phenotypes. Altogether, our analyses illustrate tissue-specific effects of Egf stimulation and highlight potential links between underlying tissue biology and Egfr signaling output.
Collapse
Affiliation(s)
- Beatrice W. Awasthi
- Center for Systems Biology, Department of Radiation Oncology, and Center for Cancer Research, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - João A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Deborah L. Burkhart
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Ian R. Smith
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Ryan L. Collins
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Cancer Program, Broad Institute of M.I.T. and Harvard, Cambridge, MA 02115, USA
| | - J. Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Kevin M. Haigis
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
36
|
Liu C, Tian X, Wang Z, Mak JCW, Mao S, Liu TM, Zheng Y. Hydrogen-induced disruption of the airway mucus barrier enhances nebulized RNA delivery to reverse pulmonary fibrosis. SCIENCE ADVANCES 2025; 11:eadt2752. [PMID: 40238879 PMCID: PMC12002117 DOI: 10.1126/sciadv.adt2752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 03/07/2025] [Indexed: 04/18/2025]
Abstract
Nebulized RNA therapies are well suited for treating respiratory diseases, in particular pulmonary fibrosis (PF); however, effective delivery remains challenging. In this study, we present a highly efficient aerosol inhalation system that enables high levels of in vivo transfection efficiency in lung macrophages, yielding durable responses against PF. First, we established a nose-only aerosol inhalation device integrated with a hydrogen supplement system. This setup enables the precise administration of lipid nanoparticles (LNPs) at a controlled low dose, while simultaneously delivering the optimal concentration of therapeutic hydrogen gas. We further developed a hybrid lipid NP (HNP) by hybridizing a pH-dependent charge-inverting lipid film with apoptotic T cell membranes to enhance endosomal escape and trigger macrophage production of hepatocyte growth factor for lung repair. We demonstrated that the hydrogen flow-induced shear stresses disrupt the NP-mucus interaction, enhancing the deposition of aerosolized HNPs/TGFβ1 siRNA within fibrotic lung lesions, effectively blocking fibrogenic signaling pathways and offering a clinically viable strategy for combating PF.
Collapse
Affiliation(s)
- Chang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xidong Tian
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Zhenping Wang
- Department of Dermatology, School of Medicine, University of California, San Diego, CA 92093, USA
| | - Judith Choi Wo Mak
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Shirui Mao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Tzu-Ming Liu
- Institute of Translational Medicine, Faculty of Health Sciences & Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau 999078, China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau 999078, China
| |
Collapse
|
37
|
Zhang H, Zhuang Z, Hong L, Wang R, Xu J, Tang Y. The malignant signature gene of cancer-associated fibroblasts serves as a potential prognostic biomarker for colon adenocarcinoma patients. Front Immunol 2025; 16:1589678. [PMID: 40313961 PMCID: PMC12043632 DOI: 10.3389/fimmu.2025.1589678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/01/2025] [Indexed: 05/03/2025] Open
Abstract
Background Colon adenocarcinoma (COAD) is the most frequently occurring type of colon cancer. Cancer-associated fibroblasts (CAFs) are pivotal in facilitating tumor growth and metastasis; however, their specific role in COAD is not yet fully understood. This research utilizes single-cell RNA sequencing (scRNA-seq) to identify and validate gene markers linked to the malignancy of CAFs. Methods ScRNA-seq data was downloaded from a database and subjected to quality control, dimensionality reduction, clustering, cell annotation, cell communication analysis, and enrichment analysis, specifically focusing on fibroblasts in tumor tissues compared to normal tissues. Fibroblast subsets were isolated, dimensionally reduced, and clustered, then combined with copy number variation (CNV) inference and pseudotime trajectory analysis to identify genes related to malignancy. A Cox regression model was constructed based on these genes, incorporating LASSO analysis, nomogram construction, and validation.Subsequently, we established two FNDC5-knockdown cell lines and utilized colony formation and transwell assays to investigate the impact of FNDC5 on cellular biological behaviors. Results Using scRNA-seq data, we analyzed 8,911 cells from normal and tumor samples, identifying six distinct cell types. Cell communication analysis highlighted interactions between these cell types mediated by ligands and receptors. CNV analysis classified CAFs into three groups based on malignancy levels. Pseudo-time analysis identified 622 pseudotime-related genes and generated a forest plot using univariate Cox regression. Lasso regression identified the independent prognostic gene FNDC5, which was visualized in a nomogram. Kaplan-Meier survival analysis confirmed the prognostic value of FNDC5, showing associations with T stage and distant metastasis. In vitro experiment results demonstrated a strong association between FNDC5 expression levels and the proliferative, migratory, and invasive abilities of colon cancer cells. Conclusion We developed a risk model for genes related to the malignancy of CAFs and identified FNDC5 as a potential therapeutic target for COAD.
Collapse
Affiliation(s)
| | | | | | | | | | - Youyuan Tang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
38
|
Huang YH, Watanabe M, Yamashita T, Sudo R. Construction of highly vascularized hepatic spheroids of primary hepatocytes via pro-angiogenic strategy in vitro. Biofabrication 2025; 17:035001. [PMID: 40179891 DOI: 10.1088/1758-5090/adc8d4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 04/03/2025] [Indexed: 04/05/2025]
Abstract
Primary hepatocytes are widely recognized for their ability to accurately represent thein vivohepatocyte phenotype. However, traditional avascular primary hepatocyte culture models are limited by inadequate mass transfer, which leads to a rapid decline in hepatocyte function and survival. To address these challenges, vascularization of hepatic spheroids is crucial for enhancing oxygen and nutrient supply, thereby enabling the construction of larger and more complex hepatic tissuesin vitro. In this study, we achieved vascularization of hepatic spheroids containing freshly isolated primary hepatocytes by incorporating fibroblasts as a source of paracrine factors to induce angiogenesis. Multicellular spheroids composed of primary hepatocytes and fibroblasts were formed in non-adhesive concave wells, and one of the spheroids was subsequently embedded in a fibrin-collagen hydrogel within a microfluidic device. Endothelial cells were then seeded onto adjacent microfluidic channels. They formed microvascular networks that extended toward and penetrated the hepatic spheroid. The vascularized hepatic spheroid closely mimicked hepatic sinusoids, with hepatocytes in close contact with microvessels. Moreover, the vascularized spheroid exhibited significantly enhanced hepatic function, specifically albumin secretion and urea synthesis. Our findings provide insights into the establishment of highly vascularized hepatic spheroidsin vitro, which is crucial for constructing scalable hepatic tissues in the context of biofabrication.
Collapse
Affiliation(s)
- Yen-Hsiang Huang
- School of Integrated Design Engineering, Graduate School of Science and Technology, Keio University, Yokohama 223-8522, Japan
| | - Masafumi Watanabe
- School of Integrated Design Engineering, Graduate School of Science and Technology, Keio University, Yokohama 223-8522, Japan
- Department of System Design Engineering, Keio University, Yokohama 223-8522, Japan
| | - Tadahiro Yamashita
- School of Integrated Design Engineering, Graduate School of Science and Technology, Keio University, Yokohama 223-8522, Japan
- Department of System Design Engineering, Keio University, Yokohama 223-8522, Japan
| | - Ryo Sudo
- School of Integrated Design Engineering, Graduate School of Science and Technology, Keio University, Yokohama 223-8522, Japan
- Department of System Design Engineering, Keio University, Yokohama 223-8522, Japan
| |
Collapse
|
39
|
Gattu AK, Tanzer M, Yaron-Barir TM, Johnson JL, Jayavelu AK, Pan H, Dreyfuss JM, Cantley LC, Mann M, Kahn CR. Cell-intrinsic insulin signaling defects in human iPS cell-derived hepatocytes in type 2 diabetes. J Clin Invest 2025; 135:e183513. [PMID: 40231468 PMCID: PMC11996863 DOI: 10.1172/jci183513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 02/07/2025] [Indexed: 04/16/2025] Open
Abstract
Hepatic insulin resistance is central to type 2 diabetes (T2D) and metabolic syndrome, but defining the molecular basis of this defect in humans is challenging because of limited tissue access. Utilizing inducible pluripotent stem cells differentiated into hepatocytes from control individuals and patients with T2D and liquid chromatography with tandem mass spectrometry-based (LC-MS/MS-based) phosphoproteomics analysis, we identified a large network of cell-intrinsic alterations in signaling in T2D. Over 300 phosphosites showed impaired or reduced insulin signaling, including losses in the classical insulin-stimulated PI3K/AKT cascade and their downstream targets. In addition, we identified over 500 phosphosites of emergent, i.e., new or enhanced, signaling. These occurred on proteins involved in the Rho-GTPase pathway, RNA metabolism, vesicle trafficking, and chromatin modification. Kinome analysis indicated that the impaired phosphorylation sites represented reduced actions of AKT2/3, PKCθ, CHK2, PHKG2, and/or STK32C kinases. By contrast, the emergent phosphorylation sites were predicted to be mediated by increased action of the Rho-associated kinases 1 and 2 (ROCK1/2), mammalian STE20-like protein kinase 4 (MST4), and/or branched-chain α-ketoacid dehydrogenase kinase (BCKDK). Inhibiting ROCK1/2 activity in T2D induced pluripotent stem cell-derived hepatocytes restored some of the alterations in insulin action. Thus, insulin resistance in the liver in T2D did not simply involve a loss of canonical insulin signaling but the also appearance of new phosphorylations representing a change in the balance of multiple kinases. Together, these led to altered insulin action in the liver and identified important targets for the therapy of hepatic insulin resistance.
Collapse
Affiliation(s)
- Arijeet K. Gattu
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, and
- Metabolism Unit and Division of Endocrinology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Maria Tanzer
- Department of Proteomics and Signal Transduction, Max Plank Institute of Biochemistry, Martinsried, Germany
- Advanced Technology and Biology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Tomer M. Yaron-Barir
- Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | | | - Ashok Kumar Jayavelu
- Department of Proteomics and Signal Transduction, Max Plank Institute of Biochemistry, Martinsried, Germany
| | - Hui Pan
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan M. Dreyfuss
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Plank Institute of Biochemistry, Martinsried, Germany
| | - C. Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, and
| |
Collapse
|
40
|
Lu J, Xiao Y, Wang Q, Chen F, Zeng Z, Yan J, Li Q, Tong Q. Development and verification of a radiomics model to forecast Ki67 index and prognosis in advanced gastric tubular adenocarcinoma. BMC Gastroenterol 2025; 25:260. [PMID: 40234767 PMCID: PMC12001714 DOI: 10.1186/s12876-025-03845-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/03/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND The purpose of this research is to evaluate the predictive capabilities of a radiomics model for the Ki67 index and its correlation with prognosis in advanced gastric tubular adenocarcinoma patients. METHODS Clinical data from 213 patients were analyzed, categorizing patients into high and low Ki67 index groups. The radiomic features of 192 patients were selected by lasso method and the model was constructed, which was validated using the TCIA dataset. Univariate and multivariate Cox regression analyses were used to further analyze clinical features associated with the prognosis of gastric cancer, radiomic models are also used to assess patient outcomes. RESULTS The radiomics model demonstrated moderate accuracy, with AUC values of 0.634, 0.666, and 0.602 for the training, validation 1, and validation 2 sets, respectively. Additionally, a significant correlation was found between the Ki67 index and radiomics scores, a higher Ki67 index was associated with improved outcomes. Kaplan-Meier analysis showed distinct survival differences between patients with high and low radiomics scores, indicating that higher scores predict better prognosis. CONCLUSIONS The radiomics model accurately predicts the Ki67 index and correlates with prognosis in advanced gastric tubular adenocarcinoma, offering valuable insights for clinical decision-making and personalized treatment strategies.
Collapse
Affiliation(s)
- Jiatong Lu
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, Hubei, China
| | - Yong Xiao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qiushuang Wang
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, Hubei, China
| | - Fangfang Chen
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhi Zeng
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Junfeng Yan
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, Hubei, China
| | - Qiang Li
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, Hubei, China
| | - Qiang Tong
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, Hubei, China.
| |
Collapse
|
41
|
Zhang Y, Fan Y, Cheng F, Chen D, Zhang H. Identification of signature genes and subtypes for heart failure diagnosis based on machine learning. Front Cardiovasc Med 2025; 12:1492192. [PMID: 40297163 PMCID: PMC12034685 DOI: 10.3389/fcvm.2025.1492192] [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: 09/06/2024] [Accepted: 03/20/2025] [Indexed: 04/30/2025] Open
Abstract
Background Heart failure (HF) is a multifaceted clinical condition, and our comprehension of its genetic pathogenesis continues to be significantly limited. Consequently, identifying specific genes for HF at the transcriptomic level may enhance early detection and allow for more targeted therapies for these individuals. Methods HF datasets were acquired from the Gene Expression Omnibus (GEO) database (GSE57338), and through the application of bioinformatics and machine-learning algorithms. We identified four candidate genes (FCN3, MNS1, SMOC2, and FREM1) that may serve as potential diagnostics for HF. Furthermore, we validated the diagnostic value of these genes on additional GEO datasets (GSE21610 and GSE76701). In addition, we assessed the different subtypes of heart failure through unsupervised clustering, and investigations were conducted on the differences in the immunological microenvironment, improved functions, and pathways among these subtypes. Finally, a comprehensive analysis of the expression profile, prognostic value, and genetic and epigenetic alterations of four potential diagnostic candidate genes was performed based on The Cancer Genome Atlas pan-cancer database. Results A total of 295 differential genes were identified in the HF dataset, and intersected with the blue module gene with the highest correlation to HF identified by weighted correlation network analysis (r = 0.72, p = 1.3 × 10-43), resulting in a total of 114 key HF genes. Furthermore, based on random forest, least absolute shrinkage and selection operator, and support vector machine algorithms, we finally identified four hub genes (FCN3, FREM1, MNS1, and SMOC2) that had good potential for diagnosis in HF (area under the curve > 0.7). Meanwhile, three subgroups for patients with HF were identified (C1, C2, and C3). Compared with the C1 and C2 groups, we eventually identified C3 as an immune subtype. Moreover, the pan-cancer study revealed that these four genes are closely associated with tumor development. Conclusions Our research identified four unique genes (FCN3, FREM1, MNS1, and SMOC2), enhancing our comprehension of the causes of HF. This provides new diagnostic insights and potentially establishes a tailored approach for individualized HF treatment.
Collapse
Affiliation(s)
- Yanlong Zhang
- Department of Cardiology, Xingtai People's Hospital, Xingtai, Hebei, China
| | - Yanming Fan
- Department of Cardiology, Xingtai People's Hospital, Xingtai, Hebei, China
| | - Fei Cheng
- Department of Cardiology, Xingtai People's Hospital, Xingtai, Hebei, China
| | - Dan Chen
- Department of Pediatrics Hematology and Oncology, Xingtai People's Hospital, Xingtai, Hebei, China
| | - Hualong Zhang
- Department of Cardiology, Xingtai People's Hospital, Xingtai, Hebei, China
| |
Collapse
|
42
|
Berasain C. New Insights and Open Questions on the Molecular and Cellular Crosstalk Governing Hepatocyte Proliferation. Cell Mol Gastroenterol Hepatol 2025:101509. [PMID: 40245923 DOI: 10.1016/j.jcmgh.2025.101509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Accepted: 03/20/2025] [Indexed: 04/19/2025]
Affiliation(s)
- Carmen Berasain
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Carlos III Health Institute), Madrid, Spain.
| |
Collapse
|
43
|
Derbeneva D, Pilmane M, Petersons A. Gene proteins, growth factors/their receptors in the wall of chronic calculous cholecystitis-affected gallbladder children. BMC Pediatr 2025; 25:288. [PMID: 40221697 PMCID: PMC11992698 DOI: 10.1186/s12887-025-05650-4] [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: 01/01/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Chronic calculous cholecystitis is the main cause of cholecystectomies in children, and 50.5% of patients with gallstones are asymptomatic at the time of diagnosis. However, the morphopathogenesis of chronic cholecystitis with cholelithiasis is unclear and may involve various genes, gene proteins, and growth factors. METHODS Tissues were obtained from four males (aged 6-18 years) and two females (aged 15 and 14 years) during planned cholecystectomies. Five healthy gallbladder tissues were obtained from the archival postmortem tissue of children. SHH, IHH, HGF, IGF1, IGF1R, and HOXB3 were detected by immunohistochemistry and evaluated semiquantitatively. Statistical analysis was used to identify statistically significant differences and correlations between the factors. RESULTS Decreased numbers of SHH-, IHH-, and IGF1R-positive cells, along with an increased number of HOXB3-positive cells, were observed in patients. SHH-positive epitheliocytes and connective tissue cells; IHH-positive cells in all locations; IGF1R-positive epitheliocytes, endotheliocytes, and smooth muscle cells; and HOXB3-positive smooth muscle cells were significantly different among the groups. However, the strongest negative correlation was found between HOXB3-positive smooth myocytes and SHH- and IHH-positive connective tissues, and the strongest positive correlation was detected among epithelial IHH, SHH, and IGF1R, as well as between IGF1R in the epithelium and endothelium of the blood vessels. CONCLUSIONS The reduced number of cells positive for the primary endodermal proteins SHH/IHH and the decreased number of IGFR1-positive cells suggest their potential roles in the development of chronic calculous cholecystitis. Additionally, the increased number of HOXB3-positive cells under these conditions likely implies stimulated growth properties, whereas HGF and IGF1 appear to have a reduced contribution to the pathogenesis of chronic calculous cholecystitis.
Collapse
Affiliation(s)
- Darja Derbeneva
- Institute of Anatomy and Anthropology, Riga Stradins University, Kronvalda Boulevard 9, Riga, LV-1010, Latvia.
| | - Mara Pilmane
- Institute of Anatomy and Anthropology, Riga Stradins University, Kronvalda Boulevard 9, Riga, LV-1010, Latvia.
| | - Aigars Petersons
- Department of Paediatric Surgery, Riga Stradins University, Dzirciema street 16, Riga, LV-1007, Latvia
| |
Collapse
|
44
|
Liu W, Tao H, Zeng X, Huang C, Yang J, Fang C. Synchronous Portal Vein Embolization of Terminal Branches with Transcatheter Arterial Chemoembolization Prior to Planned Major Hepatectomy for Hepatitis B Virus-Related Hepatocellular Carcinoma: A Single Center Retrospective Cohort Study. J Gastrointest Surg 2025:102036. [PMID: 40222551 DOI: 10.1016/j.gassur.2025.102036] [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: 12/26/2024] [Revised: 03/10/2025] [Accepted: 03/22/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Terminal branches portal vein embolization (TBPVE) is a novel technical modification. The clinical impact of synchronous TBPVE with transcatheter arterial chemoembolization (TACE) before planned major hepatectomy in hepatitis B virus (HBV) related HCC is still unknown. METHOD From November 2016 to December 2021, the study enrolled 115 patients with HBV-related HCC who were scheduled for major hepatectomy but had insufficient FLR. Patients were grouped according to whether they received TBPVE combined with TACE (the TBPVE group, n = 62) or PVE combined with TACE (the PVE group, n = 53). We compared the outcomes of the procedures and the perioperative and long-term outcomes of patients who subsequently underwent major hepatectomy between the groups. RESULTS FLR volume increment and degree of hypertrophy were significantly higher in the TBPVE group than in the PVE group (P≤0.003). FLR proliferation was pronounced remarkably within the first two weeks after TBPVE synchronous with TACE, the kinetic growth rates were 17.4 (12.7-21.9) mL/day and 6.9 (4.1-11.1) mL/day respectively. 54 patients in the TBPVE group and 41 patients in the PVE group finally underwent major hepatectomies. Pringle's maneuver time, intraoperative blood loss, transfusion, and postoperative hospital stays were less in the TBPVE group (P<0.05). Disease-free survival (DFS) rates were better in the TBPVE group than in the PVE group (P=0.042). CONCLUSION TBPVE synchronous with TACE induced safe and rapid FLR hypertrophy with satisfactory efficacy in initially unresectable HBV-related HCC patients improved the planned major hepatectomy resectability rate and has better DFS.
Collapse
Affiliation(s)
- Wenying Liu
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Department of Hepatobiliary Surgery, Yuebei People's Hospital, Shantou University Medical College, Shaoguang 512025, China
| | - Haisu Tao
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou 510280, China; National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments - South China Institute, Guangzhou 510280, China; Digital Intelligent Minimally Invasive Surgery Institute, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiaojun Zeng
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou 510280, China; National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments - South China Institute, Guangzhou 510280, China; Digital Intelligent Minimally Invasive Surgery Institute, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Congyun Huang
- Department of Hepatobiliary Surgery, Yuebei People's Hospital, Shantou University Medical College, Shaoguang 512025, China
| | - Jian Yang
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou 510280, China; National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments - South China Institute, Guangzhou 510280, China; Digital Intelligent Minimally Invasive Surgery Institute, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chihua Fang
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou 510280, China; National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments - South China Institute, Guangzhou 510280, China; Digital Intelligent Minimally Invasive Surgery Institute, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
45
|
Rodimova S, Gubarkova E, Bobrov N, Shchechkin I, Kozlova V, Zolotova N, Potapov A, Kiseleva E, Gelikonov G, Gladkova N, Zagainov V, Zagaynova E, Kuznetsova D. Optical Coherence Tomography Angiography, Elastography, and Attenuation Imaging for Evaluation of Liver Regeneration. Diagnostics (Basel) 2025; 15:977. [PMID: 40310384 PMCID: PMC12025902 DOI: 10.3390/diagnostics15080977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
Background/Objectives: As a result of metabolic changes and the disruption of tissue architecture and microcirculation, the regenerative potential of the liver decreases with violations at both micro and macro levels. The development of intraoperative approaches for assessing its regenerative potential is important for reducing the risk of the occurrence of post-resection liver failure. In this study, we used multimodal optical coherence tomography (MM OCT), a combination of three optical coherence tomography modalities-OCT-angiography (OCTA), attenuation coefficient mapping, and OCT-elastography (OCE) to provide real-time three-dimensional and label-free assessment of changes in microcirculation, and in the structure and stiffness of the liver during regeneration. Methods: In our study, the regeneration of a healthy liver was induced by 70% partial hepatectomy. Monitoring of changes was carried out on the 0 (normal liver), 3rd and 7th day of regeneration using modalities of MM OCT. OCT offers the benefits of higher resolution and specificity compared with other clinical imaging modalities, and can be used, even intraoperatively. Results: By the 3rd day of liver regeneration, a decreased density of all observable vessels, together with increased values of the liver tissue's attenuation coefficient and stiffness, was revealed compared to their initial state. However, by the 7th day, the studied parameters tended to return to their normal values, except that the density of large-caliber vessels continued to increase further. Histological and biochemical blood analysis methods were used to verify the MM OCT data. Conclusions: Such data are a first step towards further investigation of liver regeneration in pathology, and, taken in perspective, this should serve as a basis for predictive intraoperative assessment of the regenerative potential of the liver in a clinical setting.
Collapse
Affiliation(s)
- Svetlana Rodimova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky sq., 603000 Nizhny Novgorod, Russia; (E.G.); (D.K.)
| | - Ekaterina Gubarkova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky sq., 603000 Nizhny Novgorod, Russia; (E.G.); (D.K.)
| | - Nikolai Bobrov
- The Volga District Medical Centre of Federal Medical and Biological Agency, 14 Ilinskaya St., 603000 Nizhny Novgorod, Russia
| | - Ilya Shchechkin
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky sq., 603000 Nizhny Novgorod, Russia; (E.G.); (D.K.)
- Institute of Biology and Biomedicine, N.I. Lobachevsky Nizhny Novgorod National Research State University, 23 Gagarina Ave., 603022 Nizhny Novgorod, Russia
| | - Vera Kozlova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky sq., 603000 Nizhny Novgorod, Russia; (E.G.); (D.K.)
- The Volga District Medical Centre of Federal Medical and Biological Agency, 14 Ilinskaya St., 603000 Nizhny Novgorod, Russia
| | - Natalia Zolotova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky sq., 603000 Nizhny Novgorod, Russia; (E.G.); (D.K.)
| | - Arseniy Potapov
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky sq., 603000 Nizhny Novgorod, Russia; (E.G.); (D.K.)
| | - Elena Kiseleva
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky sq., 603000 Nizhny Novgorod, Russia; (E.G.); (D.K.)
| | - Grigory Gelikonov
- A.V. Gaponov-Grekhov Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanova Street, 603950 Nizhny Novgorod, Russia
| | - Natalia Gladkova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky sq., 603000 Nizhny Novgorod, Russia; (E.G.); (D.K.)
| | - Vladimir Zagainov
- Nizhny Novgorod Regional Clinical Oncologic Dispensary, Delovaya St., 11/1, 603126 Nizhny Novgorod, Russia
| | - Elena Zagaynova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky sq., 603000 Nizhny Novgorod, Russia; (E.G.); (D.K.)
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya St., 119435 Moscow, Russia
| | - Daria Kuznetsova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky sq., 603000 Nizhny Novgorod, Russia; (E.G.); (D.K.)
- Laboratory of Omics and Regenerative Technologies, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., 119991 Moscow, Russia
| |
Collapse
|
46
|
Yang Y, Wang TY, Li Q, Lu J, Ren Y, Weiner AB, Fry J, Liu Q, Yum C, Wang R, Guo Q, Wan Y, Ji Z, Dong X, Lotan TL, Schaeffer EM, Yang R, Cao Q. Androgen receptor-regulated lncRNA PRCAT71 promotes AR signaling through the interaction with KHSRP in prostate cancer. SCIENCE ADVANCES 2025; 11:eadk6989. [PMID: 40203114 PMCID: PMC11980854 DOI: 10.1126/sciadv.adk6989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Mounting evidence indicates that long noncoding RNAs (lncRNAs) play vital roles in tumorigenesis and progression of cancers. However, the functions and regulatory mechanisms of lncRNAs in prostate cancer (PCa) are still largely unknown. In this study, we found an lncRNA, PCa-associated transcript 71 (PRCAT71), highly expressed in metastatic and primary PCa compared to benign prostate tissues. Silencing PRCAT71 inhibited cancerous properties of PCa cells and androgen receptor (AR) signaling. Mechanistically, PRCAT71 acts as a scaffold to recruit K homology (KH)-type splicing regulatory protein (KHSRP) to AR messenger RNA (mRNA) and stabilize AR mRNA, leading to activated AR signaling. KHSRP plays a critical role in PCa progression. PRCAT71 is transcriptionally regulated by AR-driven enhancers, forming a positive regulatory loop between AR and PRCAT71 in PCa. Our study demonstrates a coordinated regulation of AR mRNA by lncRNA PRCAT71 and RNA binding protein KHSRP and provides insight that the PRCAT71-KHSRP-AR axis is a promising therapeutic target for treating PCa.
Collapse
Affiliation(s)
- Yongyong Yang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ting-You Wang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qianru Li
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jiawen Lu
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yanan Ren
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Adam B. Weiner
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Joshua Fry
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qi Liu
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Chaehyun Yum
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rui Wang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qingxiang Guo
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yu Wan
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Evanston, IL 60628, USA
| | - Zhe Ji
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Evanston, IL 60628, USA
| | - Xuesen Dong
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Tamara L. Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Edward M. Schaeffer
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rendong Yang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qi Cao
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
47
|
Wu C, Xie X, Yang X, Du M, Lin H, Huang J. Applications of gene pair methods in clinical research: advancing precision medicine. MOLECULAR BIOMEDICINE 2025; 6:22. [PMID: 40202606 PMCID: PMC11982013 DOI: 10.1186/s43556-025-00263-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: 09/06/2024] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
The rapid evolution of high-throughput sequencing technologies has revolutionized biomedical research, producing vast amounts of gene expression data that hold immense potential for biological discovery and clinical applications. Effectively mining these large-scale, high-dimensional data is crucial for facilitating disease detection, subtype differentiation, and understanding the molecular mechanisms underlying disease progression. However, the conventional paradigm of single-gene profiling, measuring absolute expression levels of individual genes, faces critical limitations in clinical implementation. These include vulnerability to batch effects and platform-dependent normalization requirements. In contrast, emerging approaches analyzing relative expression relationships between gene pairs demonstrate unique advantages. By focusing on binary comparisons of two genes' expression magnitudes, these methods inherently normalize experimental variations while capturing biologically stable interaction patterns. In this review, we systematically evaluate gene pair-based analytical frameworks. We classify eleven computational approaches into two fundamental categories: expression value-based methods quantifying differential expression patterns, and rank-based methods exploiting transcriptional ordering relationships. To bridge methodological development with practical implementation, we establish a reproducible analytical pipeline incorporating feature selection, classifier construction, and model evaluation modules using real-world benchmark datasets from pulmonary tuberculosis studies. These findings position gene pair analysis as a transformative paradigm for mining high-dimensional omics data, with direct implications for precision biomarker discovery and mechanistic studies of disease progression.
Collapse
Affiliation(s)
- Changchun Wu
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xueqin Xie
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xin Yang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Mengze Du
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, 611844, China
| | - Hao Lin
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Jian Huang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| |
Collapse
|
48
|
Duță C, Dogaru CB, Muscurel C, Stoian I. Nanozymes: Innovative Therapeutics in the Battle Against Neurodegenerative Diseases. Int J Mol Sci 2025; 26:3522. [PMID: 40332015 PMCID: PMC12026839 DOI: 10.3390/ijms26083522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS) and Huntington's disease (HD), represent a significant challenge to global health due to their progressive nature and the absence of curative treatments. These disorders are characterized by oxidative stress, protein misfolding, and neuroinflammation, which collectively contribute to neuronal damage and death. Recent advancements in nanotechnology have introduced nanozymes-engineered nanomaterials that mimic enzyme-like activities-as promising therapeutic agents. This review explores the multifaceted roles of nanozymes in combating oxidative stress and inflammation in neurodegenerative conditions. By harnessing their potent antioxidant properties, nanozymes can effectively scavenge reactive oxygen species (ROS) and restore redox balance, thereby protecting neuronal function. Their ability to modify surface properties enhances targeted delivery and biocompatibility, making them suitable for various biomedical applications. In this review, we highlight recent findings on the design, functionality, and therapeutic potential of nanozymes, emphasizing their dual role in addressing oxidative stress and pathological features such as protein aggregation. This synthesis of current research underscores the innovative potential of nanozymes as a proactive therapeutic strategy to halt disease progression and improve patient outcomes in neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | - Corina Muscurel
- Department of Biochemistry, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.D.); (C.B.D.); (I.S.)
| | | |
Collapse
|
49
|
Junrungsee S, Vipudhamorn W, Lapisatepun W, Thepbunchonchai A, Chotirosniramit A, Lapisatepun W, Ko-Iam W. Portal flow modulation by splenic artery ligation to prevent posthepatectomy liver failure: A randomized controlled trial. Surgery 2025:109351. [PMID: 40204604 DOI: 10.1016/j.surg.2025.109351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/27/2025] [Accepted: 03/14/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Posthepatectomy liver failure is a serious clinical issue with high mortality, similar in pathophysiology to small-for-size syndrome seen in liver transplantation. This study evaluates the efficacy of splenic artery ligation in reducing posthepatectomy liver failure in patients with portal venous pressure >15 mm Hg after hepatectomy. METHODS This single-center, randomized controlled trial was conducted from May 2019 to November 2023. Eligible participants were patients scheduled for open hepatectomy for any indication. Patients with a portal venous pressure >15 mm Hg were randomized into splenic artery ligation and control groups in a 1:1 ratio. The primary outcomes were posthepatectomy liver failure grades B and C (International Study group of Liver Surgery criteria), and secondary outcomes included 90-day mortality, comprehensive complication index, and ascites volume. RESULTS The study was terminated early, before reaching the calculated sample size, because the primary outcome in the intervention group demonstrated statistically significant results. Of the 92 patients, 36 had elevated portal venous pressure, which was associated with greater rates of posthepatectomy liver failure grades B and C (41.67% vs 3.57%, P < .001), increased ascites volume (5,340 mL vs 1,055 mL, P < .001), and a greater comprehensive complication index (20.90 vs 8.70, P < .001). In the randomized subset, splenic artery ligation significantly reduced portal venous pressure and the portal venous pressure-central venous pressure gradient compared with both presplenic artery ligation values and the control group and significantly lowered the incidence of posthepatectomy liver failure grades B and C (16.67% vs 66.67%, P = .006), comprehensive complication index (8.70 vs 20.90, P = .034). Splenic artery ligation was identified as an independent factor in reducing posthepatectomy liver failure (adjusted relative risk, 0.29). CONCLUSION Splenic artery ligation is effective in reducing posthepatectomy liver failure in patients with high portal venous pressure after hepatectomy.
Collapse
Affiliation(s)
- Sunhawit Junrungsee
- Division of Hepatobiliary Pancreatic Surgery, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Clinical Surgical Research Center, Chiang Mai University, Chiang Mai, Thailand.
| | - Witcha Vipudhamorn
- Division of Colorectal Surgery, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Worakitti Lapisatepun
- Division of Hepatobiliary Pancreatic Surgery, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand. https://twitter.com/WallEe19880
| | - Asara Thepbunchonchai
- Division of Hepatobiliary Pancreatic Surgery, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand. https://twitter.com/AsaraOum
| | - Anon Chotirosniramit
- Division of Hepatobiliary Pancreatic Surgery, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Warangkana Lapisatepun
- Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Wasana Ko-Iam
- Clinical Surgical Research Center, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
50
|
Chaudhary R, Weiskirchen R, Ehrlich M, Henis YI. Dual signaling pathways of TGF-β superfamily cytokines in hepatocytes: balancing liver homeostasis and disease progression. Front Pharmacol 2025; 16:1580500. [PMID: 40260391 PMCID: PMC12009898 DOI: 10.3389/fphar.2025.1580500] [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: 02/20/2025] [Accepted: 03/25/2025] [Indexed: 04/23/2025] Open
Abstract
The transforming growth factor-β (TGF-β) superfamily (TGF-β-SF) comprises over 30 cytokines, including TGF-β, activins/inhibins, bone morphogenetic proteins (BMPs), and growth differentiation factors (GDFs). These cytokines play critical roles in liver function and disease progression. Here, we discuss Smad-dependent (canonical) and non-Smad pathways activated by these cytokines in a hepatocellular context. We highlight the connection between the deregulation of these pathways or the balance between them and key hepatocellular processes (e.g., proliferation, apoptosis, and epithelial-mesenchymal transition (EMT)). We further discuss their contribution to various chronic liver conditions, such as metabolic dysfunction-associated steatotic liver disease (MASLD), metabolic dysfunction-associated steatohepatitis (MASH), and hepatocellular carcinoma (HCC). In MASLD and MASH, TGF-β signaling contributes to hepatocyte lipid accumulation, cell death and fibrosis progression through both Smad and non-Smad pathways. In HCC, TGF-β and other TGF-β-SF cytokines have a dual role, acting as tumor suppressors or promoters in early vs. advanced stages of tumor progression, respectively. Additionally, we review the involvement of non-Smad pathways in modulating hepatocyte responses to TGF-β-SF cytokines, particularly in the context of chronic liver diseases, as well as the interdependence with other key pathways (cholesterol metabolism, insulin resistance, oxidative stress and lipotoxicity) in MASLD/MASH pathogenesis. The perspectives and insights detailed in this review may assist in determining future research directions and therapeutic targets in liver conditions, including chronic liver diseases and cancer.
Collapse
Affiliation(s)
- Roohi Chaudhary
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yoav I. Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|