1
|
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
|
2
|
Aoyagi T, Goya T, Imoto K, Azuma Y, Hioki T, Kohjima M, Tanaka M, Oda Y, Ogawa Y. Two types of regenerative cell populations appear in acute liver injury. Stem Cell Reports 2025:102503. [PMID: 40345206 DOI: 10.1016/j.stemcr.2025.102503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 05/11/2025] Open
Abstract
The liver has a robust regenerative capacity. However, the mechanisms underlying this process remain unclear. Numerous studies on liver regeneration have been previously conducted using partial hepatectomy models, which may not fully represent acute liver injury with inflammation and necrosis. This is commonly observed in the majority of clinical cases. In this study, we conducted a single-cell RNA sequencing (RNA-seq) analysis of liver regeneration in acetaminophen-treated mice using publicly available data. We found that two distinct populations of regenerative cells simultaneously appeared within the same regenerative process. The two populations significantly differed in terms of cell morphology, differentiation, localization, proliferation rate, and signal response. Moreover, one of the populations was induced by contact with necrotic tissue and demonstrated a higher proliferative capacity with a dedifferentiated feature. These findings provide new insights into liver regeneration and therapeutic strategies for liver failure.
Collapse
Affiliation(s)
- Tomomi Aoyagi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Goya
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koji Imoto
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuki Azuma
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomonobu Hioki
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Motoyuki Kohjima
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Gastroenterology, NHO Kyushu Medical Center, Fukuoka, Japan
| | - Masatake Tanaka
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
3
|
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
|
4
|
Zhao H, Tao H, Gao J, Wang J, Hui G, Zhu Y, Wang J, Ding X, Dai Y. IL-6 Affects Liver Metabolic Abnormalities Caused by Silicon Exposure by Regulating the PKC/YY1 Signaling Pathway. Genes (Basel) 2025; 16:456. [PMID: 40282416 PMCID: PMC12026785 DOI: 10.3390/genes16040456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/31/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND This study aims to investigate the impact of coal dust (silicon dioxide) exposure on dyslipidemia and its underlying mechanisms, with a focus on the association between coal dust exposure and hepatic metabolic disorders. METHODS Clinical data were collected from 5433 coal mine workers to compare the incidence of dyslipidemia between the dust-exposed group and the non-exposed group. A mouse model of silicon dioxide exposure was established to observe hepatic fat accumulation and pathological changes. Liver tissue sequencing was performed to screen for key differential genes. In vitro cell experiments were utilized to identify the molecular mechanisms underlying hepatocyte metabolic abnormalities induced by silicon dioxide exposure. RESULTS Clinical data revealed that 69.2% of miners in the dust-exposed group developed dyslipidemia, which was higher than the 30.7% in the non-exposed group. Animal data showed that silicon dioxide exposure led to hepatic fat deposition and pathological damage, with the degree of injury positively correlated with exposure time. Liver sequencing identified a significant upregulation of the FMO3 (flavin monooxygenase 3) gene in mouse liver tissue following silicon dioxide exposure, accompanied by enhanced inflammatory responses. Mechanistic studies demonstrated that silicon dioxide activates Kupffer cells to secrete IL-6 (interleukin-6), which induces high expression of FMO3 in hepatocytes through the PKC/YY1 signaling pathway, thereby disrupting lipid metabolism. CONCLUSIONS Silicon dioxide exposure can promote the upregulation of FMO3 expression in hepatocytes by activating Kupffer cells to release IL-6 via the PKC/YY1 pathway, ultimately leading to lipid metabolic disorders and dyslipidemia.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Pharmacology, School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (H.Z.); (H.T.); (J.G.); (J.W.); (G.H.); (Y.Z.); (J.W.)
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan 232001, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan 232001, China
| | - Huihui Tao
- Department of Pharmacology, School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (H.Z.); (H.T.); (J.G.); (J.W.); (G.H.); (Y.Z.); (J.W.)
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan 232001, China
| | - Jian Gao
- Department of Pharmacology, School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (H.Z.); (H.T.); (J.G.); (J.W.); (G.H.); (Y.Z.); (J.W.)
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan 232001, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan 232001, China
| | - Jingjing Wang
- Department of Pharmacology, School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (H.Z.); (H.T.); (J.G.); (J.W.); (G.H.); (Y.Z.); (J.W.)
| | - Guangliang Hui
- Department of Pharmacology, School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (H.Z.); (H.T.); (J.G.); (J.W.); (G.H.); (Y.Z.); (J.W.)
| | - Ye Zhu
- Department of Pharmacology, School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (H.Z.); (H.T.); (J.G.); (J.W.); (G.H.); (Y.Z.); (J.W.)
| | - Jialin Wang
- Department of Pharmacology, School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (H.Z.); (H.T.); (J.G.); (J.W.); (G.H.); (Y.Z.); (J.W.)
| | - Xuansheng Ding
- Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China
| | - Yong Dai
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan 232001, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan 232001, China
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and Technology, Huainan 232001, China
| |
Collapse
|
5
|
de Oliveira THC, Gonçalves GKN. Liver ischemia reperfusion injury: Mechanisms, cellular pathways, and therapeutic approaches. Int Immunopharmacol 2025; 150:114299. [PMID: 39961215 DOI: 10.1016/j.intimp.2025.114299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/28/2025] [Accepted: 02/11/2025] [Indexed: 03/03/2025]
Abstract
Liver ischemia-reperfusion injury (LIRI) is a critical challenge in liver transplantation, resection, and trauma surgeries, leading to significant hepatic damage due to oxidative stress, inflammation, and mitochondrial dysfunction. This review explores the cellular and molecular mechanisms underlying LIRI, focusing on ATP depletion, mitochondrial dysfunction, and the involvement of reactive oxygen species (ROS). Inflammatory pathways, including the activation of nuclear factor-kappa B (NF-κB) and the NLRP3 inflammasome, as well as pro-inflammatory cytokines such as TNF-α and IL-1β, play a crucial role in exacerbating tissue damage. Various types of cell death, including necrosis, apoptosis, necroptosis, pyroptosis, ferroptosis and cuproptosis are also discussed. Therapeutic interventions targeting these mechanisms, such as antioxidants, anti-inflammatories, mitochondrial protectors, and signaling modulators, have shown promise in pre-clinical studies. However, translating these findings into clinical practice faces challenges due to the limitations of animal models and the complexity of human responses. Emerging therapies, such as RNA-based treatments, genetic editing, and stem cell therapies, offer potential breakthroughs in LIRI management. This review highlights the need for further research and the development of innovative therapeutic approaches to improve clinical outcomes.
Collapse
|
6
|
Liu D, Yan J, Luo T, Yang L. Association between C-reactive protein and hemoglobin in US rheumatoid arthritis patients based on NHANES data analysis. Sci Rep 2025; 15:8905. [PMID: 40087374 PMCID: PMC11909226 DOI: 10.1038/s41598-025-93720-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 03/10/2025] [Indexed: 03/17/2025] Open
Abstract
The poor prognosis of rheumatoid arthritis (RA) and its potential for complications highlight the importance of understanding factors that are associated with incidence and mortality rates. The inclusion criteria of this study were RA-related data from 1999 to 2018 in the National Health and Nutrition Examination Survey (NHANES) dataset. Based on certain screening criteria, a total of 610 subjects were recruited. The Low CRP group (< 3 mg/L) and the High CRP group (> 3 mg/L) were significantly different in gender, poverty-to-income ratio, body mass index, hypertension, hemoglobin (Hb), hematocrit, and mean corpuscular hemoglobin. KM survival result revealed that male RA patients in the Low Hb group had a significantly lower survival rate than those in the High Hb group (P < 0.0001), while female RA patients in the Low Hb group showed no statistically significant difference compared with the High Hb group (P = 0.13). Importantly, there was a nonlinear relationship between Hb and all-cause mortality in RA patients. In this study, we identified Hb as a protective factor against CRP in RA patients and also observed its association with the prognosis of RA. Consequently, regulating Hb levels might be considered to be associated with the progression of RA.
Collapse
Affiliation(s)
- Defang Liu
- Department of Traditional Chinese Medicine, The General Hospital of Western Theater Command, No. 270 Tianhui Road, Chengdu, 610083, Sichuan, China
| | - Jiao Yan
- Department of Traditional Chinese Medicine, The General Hospital of Western Theater Command, No. 270 Tianhui Road, Chengdu, 610083, Sichuan, China
| | - Ting Luo
- Department of Traditional Chinese Medicine, The General Hospital of Western Theater Command, No. 270 Tianhui Road, Chengdu, 610083, Sichuan, China
| | - Ling Yang
- Department of Traditional Chinese Medicine, The General Hospital of Western Theater Command, No. 270 Tianhui Road, Chengdu, 610083, Sichuan, China.
| |
Collapse
|
7
|
Hao J, Wang Z, Ren J, Cao S, Xie Z, Yang J, Li J, Ding W, Li J, Han Z, Yuan Y, Hai T, Ding S, Zhang MQ, Shi M. Single-cell multi-omics deciphers hepatocyte dedifferentiation and illuminates maintenance strategies. Cell Prolif 2025; 58:e13772. [PMID: 39810466 PMCID: PMC11882756 DOI: 10.1111/cpr.13772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/09/2024] [Accepted: 10/29/2024] [Indexed: 01/16/2025] Open
Abstract
Due to the similarity to human hepatocytes, porcine hepatocytes play an important role in hepatic research and drug evaluation. However, once hepatocytes were cultured in vitro, it was often prone to dedifferentiate, resulting in the loss of their characteristic features and normal functions, which impede their application in liver transplantation and hepatotoxic drugs evaluation. Up to now, this process has yet to be thoroughly investigated from the single-cell resolution and multi-omics perspective. In this study, we utilized 10× multiome technology to dissect the heterogeneity of porcine hepatocytes at different time points (Days 0, 1, 3, 5 and 7) during dedifferentiation. We comprehensively investigated cell heterogeneity, cellular dynamics, signalling pathways, potential gene targets, enhancer-driven gene regulatory networks, cell-cell communications of these cells and the conservation of mechanisms across species. We found that a series of critical signalling pathways driven by ERK, PI3K, Src and TGF-β were activated during this process, especially in the early stage of dedifferentiation. Based on these discoveries, we constructed a chemical combination targeting these pathways, which effectively inhibited the dedifferentiation of porcine hepatocytes in vitro. To validate the effectiveness of this combination, we transplanted such treated hepatocytes into FRGN mice, and the results demonstrated that these cells could effectively repopulate the liver and improve the survival of mice.
Collapse
Affiliation(s)
- Jie Hao
- School of Pharmaceutical SciencesTsinghua UniversityBeijingChina
| | - Zhenyi Wang
- MOE Key Laboratory of Bioinformatics, Beijing National Research Center for Information Science and Technology, Bioinformatics DivisionTsinghua UniversityBeijingChina
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai)Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jilong Ren
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Beijing Farm Animal Research Center, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Shenghao Cao
- Institute of Image Processing and Pattern RecognitionShanghai Jiao Tong UniversityShanghaiChina
- Key Laboratory of System Control and Information ProcessingMinistry of Education of ChinaShanghaiChina
| | - Zhongchen Xie
- School of Life SciencesTsinghua UniversityBeijingChina
| | - Jinghuan Yang
- School of Pharmaceutical SciencesTsinghua UniversityBeijingChina
| | - Jiachen Li
- State Key Laboratory of Biopharmaceutical Preparation and DeliveryInstitute of Process EngineeringChinese Academy of SciencesBeijingChina
| | - Weizhe Ding
- School of Pharmaceutical SciencesTsinghua UniversityBeijingChina
| | - Jie Li
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Beijing Farm Animal Research Center, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Zhiqiang Han
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Beijing Farm Animal Research Center, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Ye Yuan
- State Key Laboratory of Biopharmaceutical Preparation and DeliveryInstitute of Process EngineeringChinese Academy of SciencesBeijingChina
| | - Tang Hai
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Beijing Farm Animal Research Center, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Sheng Ding
- School of Pharmaceutical SciencesTsinghua UniversityBeijingChina
| | - Michael Q. Zhang
- MOE Key Laboratory of Bioinformatics, Beijing National Research Center for Information Science and Technology, Bioinformatics DivisionTsinghua UniversityBeijingChina
- Department of Biological Sciences, Center for Systems BiologyThe University of TexasRichardsonTexasUSA
| | - Minglei Shi
- MOE Key Laboratory of Bioinformatics, Beijing National Research Center for Information Science and Technology, Bioinformatics DivisionTsinghua UniversityBeijingChina
| |
Collapse
|
8
|
Lim B, Kamal A, Gomez Ramos B, Adrian Segarra JM, Ibarra IL, Dignas L, Kindinger T, Volz K, Rahbari M, Rahbari N, Poisel E, Kafetzopoulou K, Böse L, Breinig M, Heide D, Gallage S, Barragan Avila JE, Wiethoff H, Berest I, Schnabellehner S, Schneider M, Becker J, Helm D, Grimm D, Mäkinen T, Tschaharganeh DF, Heikenwalder M, Zaugg JB, Mall M. Active repression of cell fate plasticity by PROX1 safeguards hepatocyte identity and prevents liver tumorigenesis. Nat Genet 2025; 57:668-679. [PMID: 39948437 PMCID: PMC11906372 DOI: 10.1038/s41588-025-02081-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 01/08/2025] [Indexed: 02/20/2025]
Abstract
Cell fate plasticity enables development, yet unlocked plasticity is a cancer hallmark. While transcription master regulators induce lineage-specific genes to restrict plasticity, it remains unclear whether plasticity is actively suppressed by lineage-specific repressors. Here we computationally predict so-called safeguard repressors for 18 cell types that block phenotypic plasticity lifelong. We validated hepatocyte-specific candidates using reprogramming, revealing that prospero homeobox protein 1 (PROX1) enhanced hepatocyte identity by direct repression of alternative fate master regulators. In mice, Prox1 was required for efficient hepatocyte regeneration after injury and was sufficient to prevent liver tumorigenesis. In line with patient data, Prox1 depletion caused hepatocyte fate loss in vivo and enabled the transition of hepatocellular carcinoma to cholangiocarcinoma. Conversely, overexpression promoted cholangiocarcinoma to hepatocellular carcinoma transdifferentiation. Our findings provide evidence for PROX1 as a hepatocyte-specific safeguard and support a model where cell-type-specific repressors actively suppress plasticity throughout life to safeguard lineage identity and thus prevent disease.
Collapse
Affiliation(s)
- Bryce Lim
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Aryan Kamal
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Borja Gomez Ramos
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Juan M Adrian Segarra
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ignacio L Ibarra
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany
| | - Lennart Dignas
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Tim Kindinger
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kai Volz
- Cell Plasticity and Epigenetic Remodeling Helmholtz Group, DKFZ, Heidelberg, Germany
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Mohammad Rahbari
- Division of Chronic Inflammation and Cancer, DKFZ, Heidelberg, Germany
- Department of Surgery, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Nuh Rahbari
- Department of Surgery, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of General and Visceral Surgery, University of Ulm, Ulm, Germany
| | - Eric Poisel
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kanela Kafetzopoulou
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lio Böse
- Cell Plasticity and Epigenetic Remodeling Helmholtz Group, DKFZ, Heidelberg, Germany
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Marco Breinig
- Cell Plasticity and Epigenetic Remodeling Helmholtz Group, DKFZ, Heidelberg, Germany
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Danijela Heide
- Division of Chronic Inflammation and Cancer, DKFZ, Heidelberg, Germany
| | - Suchira Gallage
- Division of Chronic Inflammation and Cancer, DKFZ, Heidelberg, Germany
- Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Faculty of Medicine, University Tuebingen, Tübingen, Germany
| | | | - Hendrik Wiethoff
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Ivan Berest
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany
| | - Sarah Schnabellehner
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | | - Jonas Becker
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty and Faculty of Engineering Sciences, Heidelberg University, Center for Integrative Infectious Diseases Research (CIID), BioQuant, Heidelberg, Germany
| | - Dominic Helm
- Proteomics Core Facility, DKFZ, Heidelberg, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty and Faculty of Engineering Sciences, Heidelberg University, Center for Integrative Infectious Diseases Research (CIID), BioQuant, Heidelberg, Germany
- German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Translational Cancer Medicine Program and Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Helsinki, Finland
| | - Darjus F Tschaharganeh
- Cell Plasticity and Epigenetic Remodeling Helmholtz Group, DKFZ, Heidelberg, Germany
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, DKFZ, Heidelberg, Germany
- Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Faculty of Medicine, University Tuebingen, Tübingen, Germany
| | - Judith B Zaugg
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany.
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Moritz Mall
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany.
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
9
|
Li J, Sun B, Tan LX, Griffin N, Niknezhad SV, Yu C, Berthoin L, Cruz-Pacheco N, Mohabbat S, Sinada H, Efraim Y, Chen FYT, An L, Gaylord EA, Bahney CS, Lombaert IM, Knox SM. Rescue of non-healing, degenerative salivary glands by cholinergic-calcium signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.31.630834. [PMID: 39803569 PMCID: PMC11722244 DOI: 10.1101/2024.12.31.630834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Chronic degenerative wounds are often deemed irreparable, directing research efforts to focus predominantly on acute tissue injury regeneration while leaving endogenous repair mechanisms for chronically damaged tissues largely unexplored. In this study, we demonstrate that non-healing, severely degenerated salivary gland tissues can be fundamentally restored through first-line treatment with muscarinic agonists. This approach rescues tissue structure and function, returning it to a homeostatic-like state, and reactivates endogenous regeneration processes to drive new cell expansion that persists for months post-treatment. Furthermore, neuromimetic activation profoundly depletes radiation-induced DNA damage and re-establishes the nerve-acinar relationship, ultimately restoring the tissues physiological capacity to maintain homeostasis, even in the absence of treatment. We show that full recovery of organ function, comparable to uninjured controls, is primarily mediated by the re-differentiation of aberrantly de-differentiated epithelial acinar cells and the restoration of mitochondrial function via a muscarinic-calcium signaling pathway. These findings challenge the prevailing notion that chronic organ degeneration is irreversible and propose a readily testable therapeutic strategy for epithelial restoration with potential applications across a spectrum of chronic injuries.
Collapse
Affiliation(s)
- Jianlong Li
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
- These authors contributed equally
| | - Bo Sun
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
- These authors contributed equally
| | - Li Xuan Tan
- Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, California, USA; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
- These authors contributed equally
| | - Nathan Griffin
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Seyyed Vahid Niknezhad
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Chieh Yu
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Lionel Berthoin
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Noel Cruz-Pacheco
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Seayar Mohabbat
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Hanan Sinada
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Yael Efraim
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Feeling Yu Ting Chen
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Luye An
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Eliza A. Gaylord
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Chelsey S. Bahney
- University of California, San Francisco. Orthopedic Trauma Institute, San Francisco, CA
| | - Isabelle M.A. Lombaert
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA
- Co–senior authors
| | - Sarah M. Knox
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
- Co–senior authors
- Lead contact
| |
Collapse
|
10
|
Humphries C, Addison ML, Dear JW, Forbes SJ. The emerging role of alternatively activated macrophages to treat acute liver injury. Arch Toxicol 2025; 99:103-114. [PMID: 39503878 PMCID: PMC11742291 DOI: 10.1007/s00204-024-03892-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/09/2024] [Indexed: 01/19/2025]
Abstract
Acute liver injury (ALI) has a clear requirement for novel therapies. One emerging option is the use of alternatively activated macrophages (AAMs); a distinct subtype of macrophage with a role in liver injury control and repair. In this comprehensive review, we provide an overview of the current limited options for ALI, and the potential advantages offered by AAMs. We describe the evidence supporting their use from in vitro studies, pre-clinical animal studies, and human clinical trials. We suggest why the first evidence for the clinical use of AAMs is likely to be found in acetaminophen toxicity, and discuss the specific evidence for AAM use in this population, as well as potential applications for AAMs in other patient populations. The key domains by which the performance of AAMs for the treatment of ALI will be assessed are identified, and remaining challenges to the successful delivery of AAMs to clinic are explored.
Collapse
Affiliation(s)
- Chris Humphries
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Drive, Edinburgh, UK
| | - Melisande L Addison
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Drive, Edinburgh, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - James W Dear
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Drive, Edinburgh, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
11
|
Zhao X, Wang S, Liu Q, Wei W, Sun X, Song H, Xu J, Zhang S, Wang H, Fu J. Single-cell landscape of the intrahepatic ecosystem in alcohol-related liver disease. Clin Transl Med 2025; 15:e70198. [PMID: 39834100 PMCID: PMC11746962 DOI: 10.1002/ctm2.70198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Alcohol-related liver disease (ALD) is a common chronic liver disease caused by long-term excessive alcohol consumption and responsible for more than half of all liver-related deaths worldwide. The molecular mechanisms associated with ALD were not fully understood. In this study, we performed single-cell RNA sequencing on liver tissues obtained from ALD patients and healthy liver donors. We identified an ALB+KRT7+ epithelial population that expressed both hepatocyte and biliary markers significantly expanded in ALD livers. The ALB+KRT7+ epithelial cells were demonstrated to have stem cell properties and malignant transformation potentials. Moreover, ALB+KRT7+ epithelium-derived ALD organoids promote the tumour growth by activating Wnt/β-catenin signalling of liver cancer cells. Most importantly, blocking the Wnt protein secretion or knockdown the Wnt receptor suppressed the tumour promoting effect of ALD organoids. Our study provides important insights that Wnt signalling can be targeted in patients with advanced alcohol-related cirrhosis to prevent malignant transformation. In addition, our results also uncovered the important alterations of nonparenchymal cells, especially macrophages and T/NK populations that responsible for active inflammation responses in alcohol-related hepatitis and immunosuppressive microenvironment in advanced cirrhosis livers, which likely facilitated the malignant progression of ALD. KEY POINTS: This study provides single-cell landscape of human liver samples across different ALD stages. The ALB+ KRT7+ epithelium were enriched in ALD patients, and the function of this epithelial population varied significantly across ALD stages. ALB+KRT7+ epithelium from advanced alcohol-related cirrhosis had malignant transformation potential and tumour promotion activity. The comprehensive changes of parenchymal and nonparenchymal cells in the ALD livers lay a hidden danger for the further malignant progression.
Collapse
Affiliation(s)
- Xiaofang Zhao
- Translational Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepatobiliary Tumor BiologyEastern Hepatobiliary Surgery Hospital, Second Military Medical University/NAVAL Medical UniversityShanghaiChina
| | - Senyan Wang
- Translational Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Qi Liu
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Wenjuan Wei
- Translational Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xiaoyan Sun
- Translational Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hao Song
- Translational Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Jing Xu
- Translational Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hongyang Wang
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepatobiliary Tumor BiologyEastern Hepatobiliary Surgery Hospital, Second Military Medical University/NAVAL Medical UniversityShanghaiChina
| | - Jing Fu
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepatobiliary Tumor BiologyEastern Hepatobiliary Surgery Hospital, Second Military Medical University/NAVAL Medical UniversityShanghaiChina
| |
Collapse
|
12
|
Ko C, Cheng CC, Mistretta D, Ambike S, Sacherl J, Velkov S, Liao BH, Bester R, Gültan M, Polezhaeva O, Herrmann A, Jakwerth CA, Schmidt-Weber CB, Bugert JJ, Wölfel R, Grass V, Essbauer S, Schnepf D, Keppler OT, Vondran FWR, Pichlmair A, Mogler C, Ebert G, Protzer U. SARS-CoV-2 Productively Infects Human Hepatocytes and Induces Cell Death. J Med Virol 2025; 97:e70156. [PMID: 39760326 DOI: 10.1002/jmv.70156] [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: 07/02/2024] [Revised: 11/25/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025]
Abstract
SARS-CoV-2 infection is accompanied by elevated liver enzymes, and patients with pre-existing liver conditions experience more severe disease. While it was known that SARS-CoV-2 infects human hepatocytes, our study determines the mechanism of infection, demonstrates viral replication and spread, and highlights direct hepatocyte damage. Viral replication was readily detectable upon infection of primary human hepatocytes and hepatoma cells with the ancestral SARS-CoV-2, Delta, and Omicron variants. Hepatocytes express the SARS-CoV-2 receptor ACE2 and the host cell protease TMPRSS2, and knocking down ACE2 and TMPRSS2 impaired SARS-CoV-2 infection. Progeny viruses released from infected hepatocytes showed the typical coronavirus morphology by electron microscopy and proved infectious when transferred to fresh cells, indicating that hepatocytes can contribute to virus spread. Importantly, SARS-CoV-2 infection rapidly induced hepatocyte death in a replication-dependent fashion, with the Omicron variant showing faster onset but less extensive cell death. C57BL/6 wild-type mice infected with a mouse-adapted SARS-CoV-2 strain showed high levels of viral RNA in liver and lung tissues. ALT peaked when viral RNA was cleared from the liver. Liver histology revealed profound tissue damage and immune cell infiltration, indicating that direct cytopathic effects of SARS-CoV-2 and immune-mediated killing of infected hepatocytes contribute to liver pathology.
Collapse
Grants
- This study was supported by the German Research Foundation (DFG) via SFB-TRR179 (project 272983813 to U.P.), TRR22 (project 398577603 to C.S.W.) and TRR353 (project 471011418 to G.E.), by the State of Bavaria via research network FOR-COVID and Bay-VOC, by the project "Virological and immunological determinants of COVID-19 pathogenesis-lessons to get prepared for future pandemics" (KA1-Co-02 "COVIPA" to U.P.) and "Airborne Transmission of SARS Coronavirus - From Fundamental Science to Efficient Air Cleaning Systems" (KA1-Co-06 "CORAERO" to G.E.), grants from the Helmholtz Association's Initiative and Networking Fund, by the European Commission FET Open Grant VIROFIGHT (grant no. 899619), by the State of Bavaria and the European Union via a grant for regional infrastructure development (EFRE - REACT, to U.P. and G.E.), by the State of Bavaria via research networks FOR-COVID and Bay-VOC (to U.P. and O.T.K.) by the Federal Ministry of Education and Research (project ESCAPE; 01KI20169A to C.S.W.), and by the Medical Biological Defense Research Program of the Bundeswehr Medical Service (to J.J.B.). In addition, this research was supported by intramural funds from KRICT (project KK2432-10 and BSF24-111 to C.K.).
Collapse
Affiliation(s)
- Chunkyu Ko
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, South Korea
| | - Cho-Chin Cheng
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Daniele Mistretta
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Shubhankar Ambike
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Julia Sacherl
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Stoyan Velkov
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Bo-Hung Liao
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Romina Bester
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Merve Gültan
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Olga Polezhaeva
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Alexander Herrmann
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Constanze A Jakwerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich/Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy & Environment (ZAUM), Technical University of Munich/Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
- German Center for Lung Research (DZL), Munich Partner Site, Munich, Germany
| | - Joachim J Bugert
- Department of Viruses and Intracellular Pathogens, Bundeswehr Institute of Microbiology, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
| | - Roman Wölfel
- Department of Viruses and Intracellular Pathogens, Bundeswehr Institute of Microbiology, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
| | - Vincent Grass
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Sandra Essbauer
- Department of Viruses and Intracellular Pathogens, Bundeswehr Institute of Microbiology, Munich, Germany
| | - Daniel Schnepf
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany
- Immunoregulation Laboratory, The Francis Crick Institute, London, UK
| | - Oliver T Keppler
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
- Max von Pettenkofer Institute & Gene Center, Faculty of Medicine, University of Munich, Munich, Germany
| | - Florian W R Vondran
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
- ReMediES, Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Andreas Pichlmair
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
| | - Carolin Mogler
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Gregor Ebert
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
| |
Collapse
|
13
|
Wang G, Qiu X, Sun M, Li Y, Chen A, Tong J, Cheng Z, Zhao W, Chang C, Yu G. RegⅢγ promotes the proliferation, and inhibits inflammation response of macrophages by Akt, STAT3 and NF-κB pathways. Int Immunopharmacol 2024; 143:113442. [PMID: 39490142 DOI: 10.1016/j.intimp.2024.113442] [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: 06/22/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024]
Abstract
As an inflammatory regulator, intestinal regenerating islet-derived 3 gamma (RegⅢγ) contributes to alleviating liver injury in liver diseases and colitis. However, it is unclear whether hepatic RegⅢγ exerts a vital impact on liver regeneration (LR). In this study, the expression profile and localization of RegⅢγ in LR were demonstrated by microarray analysis, qRT-PCR and immunofluorescence staining. Then, RAW264.7 cells with RegⅢγ deficiency and overexpression were obtained by the CRISPR/Cas9 system and lentivirus infection, respectively. MTT, flow cytometry, EdU, transwell, neutral red phagocytosis, and NO assays were performed to detect the functions of RegⅢγ in RAW264.7 cell proliferation and inflammation. Finally, the regulatory mechanism of RegⅢγ was explored by co-immunoprecipitation and Western blot assays. According to our findings, RegⅢγ showed significant expression changes in Kupffer cells during LR, and RegⅢγ overexpression stimulated the viability, proliferation, phagocytosis and migration of RAW264.7 cells, whereas RegⅢγ deficiency reversed these effects. Similarly, RegⅢγ overexpression facilitated the expression of HO-1 and IL-10, while RegⅢγ deficiency promoted NO production and p-Akt, p-STAT3, p-p65 and TNF-α expression. In conclusion, RegⅢγ may facilitate LR by promoting the proliferation of macrophages and inhibiting their inflammatory response through Akt, STAT3 and NF-κB pathways in the priming stage of LR.
Collapse
Affiliation(s)
- Gaiping Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Henan Normal University, Xinxiang 453007, Henan, China; Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Normal University, Xinxiang 453007, Henan, China; Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China.
| | - Xianglei Qiu
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China
| | - Meiqing Sun
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China
| | - Yingle Li
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China
| | - Anqi Chen
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China
| | - Jiahui Tong
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China
| | - Zhipeng Cheng
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China
| | - Weiming Zhao
- Institute of Biomedical Sciences, Henan Academy of Sciences, Zhengzhou 450046, Henan, China
| | - Cuifang Chang
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Henan Normal University, Xinxiang 453007, Henan, China; Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Normal University, Xinxiang 453007, Henan, China; Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China
| | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Henan Normal University, Xinxiang 453007, Henan, China; Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Normal University, Xinxiang 453007, Henan, China; Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China.
| |
Collapse
|
14
|
Bi R, Pan LN, Dai H, Sun C, Li C, Lin HJ, Xie LP, Ma HX, Li L, Xie H, Guo K, Hou CH, Yao YG, Chen LN, Zheng P. Epigenetic characterization of adult rhesus monkey spermatogonial stem cells identifies key regulators of stem cell homeostasis. Nucleic Acids Res 2024; 52:13644-13664. [PMID: 39535033 DOI: 10.1093/nar/gkae1013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/12/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Spermatogonial stem cells (SSCs) play crucial roles in the preservation of male fertility. However, successful ex vivo expansion of authentic human SSCs remains elusive due to the inadequate understanding of SSC homeostasis regulation. Using rhesus monkeys (Macaca mulatta) as a representative model, we characterized SSCs and progenitor subsets through single-cell RNA sequencing using a cell-specific network approach. We also profiled chromatin status and major histone modifications (H3K4me1, H3K4me3, H3K27ac, H3K27me3 and H3K9me3), and subsequently mapped promoters and active enhancers in TSPAN33+ putative SSCs. Comparing the epigenetic changes between fresh TSPAN33+ cells and cultured TSPAN33+ cells (resembling progenitors), we identified the regulatory elements with higher activity in SSCs, and the potential transcription factors and signaling pathways implicated in SSC regulation. Specifically, TGF-β signaling is activated in monkey putative SSCs. We provided evidence supporting its role in promoting self-renewal of monkey SSCs in culture. Overall, this study outlines the epigenetic landscapes of monkey SSCs and provides clues for optimization of the culture condition for primate SSCs expansion.
Collapse
Affiliation(s)
- Rui Bi
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Baohua Road, Kunming 650107, China
| | - Lin-Nuo Pan
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, No. 320 Yue Yang Road, Shanghai 200031, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
| | - Hao Dai
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, No. 320 Yue Yang Road, Shanghai 200031, China
| | - Chunli Sun
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
| | - Cong Li
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
| | - Hui-Juan Lin
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Baohua Road, Kunming 650107, China
| | - Lan-Ping Xie
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
| | - Huai-Xiao Ma
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Baohua Road, Kunming 650107, China
| | - Lin Li
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
| | - Heng Xie
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
| | - Kun Guo
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
| | - Chun-Hui Hou
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
| | - Yong-Gang Yao
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Baohua Road, Kunming 650107, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
| | - Luo-Nan Chen
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, No. 320 Yue Yang Road, Shanghai 200031, China
- Key Laboratory of Systems Biology, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, No. 1 Xiangshan Branch Lane, Xihu District, Hangzhou 310024, China
| | - Ping Zheng
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Baohua Road, Kunming 650107, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
| |
Collapse
|
15
|
Dang C, Liu M, Liu P, Liu J, Yu X, Dong Y, Zhao J. Causal relationship between inflammatory factors and gynecological cancer: a Bayesian Mendelian randomization study. Sci Rep 2024; 14:29868. [PMID: 39622847 PMCID: PMC11612437 DOI: 10.1038/s41598-024-80747-x] [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: 07/11/2024] [Accepted: 11/21/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Cervical cancer, ovarian cancer, and endometrial cancer are the three most common cancers in gynecology. Understanding their respective pathology is currently incomplete. Inflammatory factors play an important role in the pathophysiology of these three cancers, but the causal relationship between inflammatory factors and these three cancers is unclear. METHODS Based on publicly available genetic databases, relevant instrumental variables were extracted according to predefined thresholds, and causal analyses of CRP, 41 circulating inflammatory factors, and three gynecological cancers were performed, mainly using the inverse variance weighted method, while bayesian analysis was performed to improve the accuracy of the results. Finally, heterogeneity, horizontal pleiotropy test, and MR Steiger test were carried out to evaluate the reliability of the findings and the causal inference strength. RESULTS One inflammatory factor (PDGF-BB) and four inflammatory factors (CXCL9, IL-6, CXCL1, and G-CSF) were identified as significantly associated with the risk of ovarian and endometrial cancers, respectively. In comparison, cervical cancer was found to have a negative causal association with one inflammatory factor (G-CSF) and endometrial cancer with two inflammatory factors (CXCL10 and CCL11). CONCLUSIONS Our MR study suggests potential causal relationships between circulating inflammatory regulators and three gynecological cancers from a genetic perspective, which contributes to further understanding of the pathomechanisms of cervical, ovarian and endometrial cancers and highlights the potential of targeting inflammatory factors as therapeutic interventions and predictors.
Collapse
Affiliation(s)
- Chunxiao Dang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Mengmeng Liu
- Dongying People's Hospital (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, 257000, China
| | - Pengfei Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Jinxing Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Xiao Yu
- Department of Gynecology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yan Dong
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingba Road No. 1, Jinan, 250014, China.
| | - Junde Zhao
- Shandong University of Traditional Chinese Medicine, Jingshi Road No. 16369, Jinan, 250014, China.
| |
Collapse
|
16
|
Wei Y, Shao J, Wei K, Peng L, Wei X. Influence of Qingzhuan Tea Polysaccharides on F - Adsorption: Molecular Structure, Binding Behavior, and In Vitro and In Vivo Digestion and Metabolism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:26384-26403. [PMID: 39545705 DOI: 10.1021/acs.jafc.4c05734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
The high level of fluoride in Qingzhuan tea (QZT) poses a potential health risk to consumers. This study aims to explore the binding behavior of purified Qingzhuan tea polysaccharides (pTPS) and fluoride ions (F-), as well as their regulatory role in the digestion and metabolism of fluoride. The sugar content of pTPS was 94.64 ± 3.01%, with a molecular weight of 7.373 × 104 Da and high homogeneity. The effects of different proportions and environmental conditions on the adsorption of F- by pTPS were investigated. The influence of the complexation of pTPS and F- on the digestion and metabolism of fluoride was explored using an in vitro gastrointestinal digestion model and C57BL/6 mice. The structural alterations of pTPS were observed during simulated gastrointestinal digestion. Furthermore, pTPS were found to reduce serum fluoride levels and inhibit accumulation in major organs and tissues, especially the heart, liver, kidneys, muscles, and femur. This study investigated the binding pattern between fluorine and pTPS and its influence on the digestion and absorption of fluorine, providing a promising potential for pTPS as a bioadsorbent of fluorine to alleviate the toxicity of fluorine in QZT, which laid a theoretical foundation for the safety of consumption of QZT.
Collapse
Affiliation(s)
- Yang Wei
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Jie Shao
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Kang Wei
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Lanlan Peng
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Xinlin Wei
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| |
Collapse
|
17
|
Huang XB, Huang Q, Jiang MC, Zhong Q, Zheng HL, Wang JB, Huang ZN, Wang HG, Liu ZY, Li YF, Xu KX, Lin M, Li P, Huang ZH, Xie JW, Lin JX, Lu J, Que JW, Zheng CH, Chen QY, Huang CM. KLHL21 suppresses gastric tumourigenesis via maintaining STAT3 signalling equilibrium in stomach homoeostasis. Gut 2024; 73:1785-1798. [PMID: 38969490 DOI: 10.1136/gutjnl-2023-331111] [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: 09/11/2023] [Accepted: 06/13/2024] [Indexed: 07/07/2024]
Abstract
OBJECTIVE Precancerous metaplasia transition to dysplasia poses a risk for subsequent intestinal-type gastric adenocarcinoma. However, the molecular basis underlying the transformation from metaplastic to cancerous cells remains poorly understood. DESIGN An integrated analysis of genes associated with metaplasia, dysplasia was conducted, verified and characterised in the gastric tissues of patients by single-cell RNA sequencing and immunostaining. Multiple mouse models, including homozygous conditional knockout Klhl21-floxed mice, were generated to investigate the role of Klhl21 deletion in stemness, DNA damage and tumour formation. Mass-spectrometry-based proteomics and ribosome sequencing were used to elucidate the underlying molecular mechanisms. RESULTS Kelch-like protein 21 (KLHL21) expression progressively decreased in metaplasia, dysplasia and cancer. Genetic deletion of Klhl21 enhances the rapid proliferation of Mist1+ cells and their descendant cells. Klhl21 loss during metaplasia facilitates the recruitment of damaged cells into the cell cycle via STAT3 signalling. Increased STAT3 activity was confirmed in cancer cells lacking KLHL21, boosting self-renewal and tumourigenicity. Mechanistically, the loss of KLHL21 promotes PIK3CB mRNA translation by stabilising the PABPC1-eIF4G complex, subsequently causing STAT3 activation. Pharmacological STAT3 inhibition by TTI-101 elicited anticancer effects, effectively impeding the transition from metaplasia to dysplasia. In patients with gastric cancer, low levels of KLHL21 had a shorter survival rate and a worse response to adjuvant chemotherapy. CONCLUSIONS Our findings highlighted that KLHL21 loss triggers STAT3 reactivation through PABPC1-mediated PIK3CB translational activation, and targeting STAT3 can reverse preneoplastic metaplasia in KLHL21-deficient stomachs.
Collapse
Affiliation(s)
- Xiao-Bo Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Qiang Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Mei-Chen Jiang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Hua-Long Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Ze-Ning Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Hua-Gen Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Zhi-Yu Liu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Yi-Fan Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Kai-Xiang Xu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Zhi-Hong Huang
- Public Technology Service Center, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Jian-Wen Que
- Columbia Center for Human Development, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian, China
| |
Collapse
|
18
|
Qin X, Tape CJ. Functional analysis of cell plasticity using single-cell technologies. Trends Cell Biol 2024; 34:854-864. [PMID: 38355348 DOI: 10.1016/j.tcb.2024.01.006] [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: 11/02/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 02/16/2024]
Abstract
Metazoan organisms are heterocellular systems composed of hundreds of different cell types, which arise from an isogenic genome through differentiation. Cellular 'plasticity' further enables cells to alter their fate in response to exogenous cues and is involved in a variety of processes, such as wound healing, infection, and cancer. Recent advances in cellular model systems, high-dimensional single-cell technologies, and lineage tracing have sparked a renaissance in plasticity research. Here, we discuss the definition of cell plasticity, evaluate state-of-the-art model systems and techniques to study cell-fate dynamics, and explore the application of single-cell technologies to obtain functional insights into cell plasticity in healthy and diseased tissues. The integration of advanced biomimetic model systems, single-cell technologies, and high-throughput perturbation studies is enabling a new era of research into non-genetic plasticity in metazoan systems.
Collapse
Affiliation(s)
- Xiao Qin
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Oxford, OX3 9DS, UK.
| | - Christopher J Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London, WC1E 6DD, UK.
| |
Collapse
|
19
|
Poss KD, Tanaka EM. Hallmarks of regeneration. Cell Stem Cell 2024; 31:1244-1261. [PMID: 39163854 PMCID: PMC11410156 DOI: 10.1016/j.stem.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/12/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024]
Abstract
Regeneration is a heroic biological process that restores tissue architecture and function in the face of day-to-day cell loss or the aftershock of injury. Capacities and mechanisms for regeneration can vary widely among species, organs, and injury contexts. Here, we describe "hallmarks" of regeneration found in diverse settings of the animal kingdom, including activation of a cell source, initiation of regenerative programs in the source, interplay with supporting cell types, and control of tissue size and function. We discuss these hallmarks with an eye toward major challenges and applications of regenerative biology.
Collapse
Affiliation(s)
- Kenneth D Poss
- Duke Regeneration Center and Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Elly M Tanaka
- Institute of Molecular Biotechnology (IMBA), Austrian Academy of Sciences, Vienna Biocenter (VBC), 1030 Vienna, Austria.
| |
Collapse
|
20
|
Wang S, Wang X, Wang Y. The Progress and Promise of Lineage Reprogramming Strategies for Liver Regeneration. Cell Mol Gastroenterol Hepatol 2024; 18:101395. [PMID: 39218152 PMCID: PMC11530608 DOI: 10.1016/j.jcmgh.2024.101395] [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: 03/15/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
The liver exhibits remarkable regenerative capacity. However, the limited ability of primary human hepatocytes to proliferate in vitro, combined with a compromised regenerative capacity induced by pathological conditions in vivo, presents significant obstacles to effective liver regeneration following liver injuries and diseases. Developing strategies to compensate for the loss of endogenous hepatocytes is crucial for overcoming these challenges, and this remains an active area of investigation. Lineage reprogramming, the process of directly converting one cell type into another bypassing the intermediate pluripotent state, has emerged as a promising method for generating specific cell types for therapeutic purposes in regenerative medicine. Here, we discuss the recent progress and emergent technologies in lineage reprogramming into hepatic cells, and their potential applications in enhancing liver regeneration or treating liver disease models. We also address controversies and challenges that confront this field.
Collapse
Affiliation(s)
- Shuyong Wang
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, China.
| | - Xuan Wang
- Hepatopancreatobiliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Beijing, China
| | - Yunfang Wang
- Hepatopancreatobiliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Beijing, China.
| |
Collapse
|
21
|
Yan L, Wang J, Cai X, Liou Y, Shen H, Hao J, Huang C, Luo G, He W. Macrophage plasticity: signaling pathways, tissue repair, and regeneration. MedComm (Beijing) 2024; 5:e658. [PMID: 39092292 PMCID: PMC11292402 DOI: 10.1002/mco2.658] [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: 03/03/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Macrophages are versatile immune cells with remarkable plasticity, enabling them to adapt to diverse tissue microenvironments and perform various functions. Traditionally categorized into classically activated (M1) and alternatively activated (M2) phenotypes, recent advances have revealed a spectrum of macrophage activation states that extend beyond this dichotomy. The complex interplay of signaling pathways, transcriptional regulators, and epigenetic modifications orchestrates macrophage polarization, allowing them to respond to various stimuli dynamically. Here, we provide a comprehensive overview of the signaling cascades governing macrophage plasticity, focusing on the roles of Toll-like receptors, signal transducer and activator of transcription proteins, nuclear receptors, and microRNAs. We also discuss the emerging concepts of macrophage metabolic reprogramming and trained immunity, contributing to their functional adaptability. Macrophage plasticity plays a pivotal role in tissue repair and regeneration, with macrophages coordinating inflammation, angiogenesis, and matrix remodeling to restore tissue homeostasis. By harnessing the potential of macrophage plasticity, novel therapeutic strategies targeting macrophage polarization could be developed for various diseases, including chronic wounds, fibrotic disorders, and inflammatory conditions. Ultimately, a deeper understanding of the molecular mechanisms underpinning macrophage plasticity will pave the way for innovative regenerative medicine and tissue engineering approaches.
Collapse
Affiliation(s)
- Lingfeng Yan
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Jue Wang
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Xin Cai
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Yih‐Cherng Liou
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
- National University of Singapore (NUS) Graduate School for Integrative Sciences and EngineeringNational University of SingaporeSingaporeSingapore
| | - Han‐Ming Shen
- Faculty of Health SciencesUniversity of MacauMacauChina
| | - Jianlei Hao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University)Jinan UniversityZhuhaiGuangdongChina
- The Biomedical Translational Research InstituteFaculty of Medical ScienceJinan UniversityGuangzhouGuangdongChina
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospitaland West China School of Basic Medical Sciences and Forensic MedicineSichuan University, and Collaborative Innovation Center for BiotherapyChengduChina
| | - Gaoxing Luo
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Weifeng He
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| |
Collapse
|
22
|
Viragova S, Li D, Klein OD. Activation of fetal-like molecular programs during regeneration in the intestine and beyond. Cell Stem Cell 2024; 31:949-960. [PMID: 38971147 PMCID: PMC11235077 DOI: 10.1016/j.stem.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/10/2024] [Accepted: 05/24/2024] [Indexed: 07/08/2024]
Abstract
Tissue regeneration after damage is generally thought to involve the mobilization of adult stem cells that divide and differentiate into progressively specialized progeny. However, recent studies indicate that tissue regeneration can be accompanied by reversion to a fetal-like state. During this process, cells at the injury site reactivate programs that operate during fetal development but are typically absent in adult homeostasis. Here, we summarize our current understanding of the molecular signals and epigenetic mediators that orchestrate "fetal-like reversion" during intestinal regeneration. We also explore evidence for this phenomenon in other organs and species and highlight open questions that merit future examination.
Collapse
Affiliation(s)
- Sara Viragova
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Dong Li
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA
| | - Ophir D Klein
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA.
| |
Collapse
|
23
|
Lee J, Gil D, Park H, Lee Y, Mun SJ, Shin Y, Jo E, Windisch MP, Kim JH, Son MJ. A multicellular liver organoid model for investigating hepatitis C virus infection and nonalcoholic fatty liver disease progression. Hepatology 2024; 80:186-201. [PMID: 37976400 DOI: 10.1097/hep.0000000000000683] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND AND AIMS HCV infection can be successfully managed with antiviral therapies; however, progression to chronic liver disease states, including NAFLD, is common. There is currently no reliable in vitro model for investigating host-viral interactions underlying the link between HCV and NAFLD; although liver organoids (LOs) show promise, they currently lack nonparenchymal cells, which are key to modeling disease progression. APPROACH AND RESULTS Here, we present a novel, multicellular LO model using a coculture system of macrophages and LOs differentiated from the same human pluripotent stem cells (PSCs). The cocultured macrophages shifted toward a Kupffer-like cell type, the liver-resident macrophages present in vivo , providing a suitable model for investigating NAFLD pathogenesis. With this multicellular Kupffer-like cell-containing LO model, we found that HCV infection led to lipid accumulation in LOs by upregulating host lipogenesis, which was more marked with macrophage coculture. Reciprocally, long-term treatment of LOs with fatty acids upregulated HCV amplification and promoted inflammation and fibrosis. Notably, in our Kupffer-like cell-containing LO model, the effects of 3 drugs for NASH that have reached phase 3 clinical trials exhibited consistent results with the clinical outcomes. CONCLUSIONS Taken together, we introduced a multicellular LO model consisting of hepatocytes, Kupffer-like cells, and HSCs, which recapitulated host-virus intercommunication and intercellular interactions. With this novel model, we present a physiologically relevant system for the investigation of NAFLD progression in patients with HCV.
Collapse
Affiliation(s)
- Jaeseo Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Dayeon Gil
- Korea National Stem Cell Bank, Chungcheongbuk-do, Republic of Korea
- Department of Chronic Diseases Convergence Research, Division of Intractable Diseases Research, Korea National Institute of Health, Osong Health Technology Administration Complex, Republic of Korea
| | - Hyeyeon Park
- Korea National Stem Cell Bank, Chungcheongbuk-do, Republic of Korea
- Department of Chronic Diseases Convergence Research, Division of Intractable Diseases Research, Korea National Institute of Health, Osong Health Technology Administration Complex, Republic of Korea
| | - Youngsun Lee
- Korea National Stem Cell Bank, Chungcheongbuk-do, Republic of Korea
- Department of Chronic Diseases Convergence Research, Division of Intractable Diseases Research, Korea National Institute of Health, Osong Health Technology Administration Complex, Republic of Korea
| | - Seon Ju Mun
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Yongbo Shin
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, Korea University of Science & Technology (UST), Daejeon, Republic of Korea
| | - Eunji Jo
- Applied Molecular Virology Laboratory, Institute Pasteur Korea, Seongnam-si, Republic of Korea
| | - Marc P Windisch
- Applied Molecular Virology Laboratory, Institute Pasteur Korea, Seongnam-si, Republic of Korea
| | - Jung-Hyun Kim
- Korea National Stem Cell Bank, Chungcheongbuk-do, Republic of Korea
- Department of Chronic Diseases Convergence Research, Division of Intractable Diseases Research, Korea National Institute of Health, Osong Health Technology Administration Complex, Republic of Korea
- College of Pharmacy, Ajou University, Suwon, Republic of Korea
| | - Myung Jin Son
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, Korea University of Science & Technology (UST), Daejeon, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
24
|
Wang MJ, Zhang HL, Chen F, Guo XJ, Liu QG, Hou J. The double-edged effects of IL-6 in liver regeneration, aging, inflammation, and diseases. Exp Hematol Oncol 2024; 13:62. [PMID: 38890694 PMCID: PMC11184755 DOI: 10.1186/s40164-024-00527-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine and exerts its complex biological functions mainly through three different signal modes, called cis-, trans-, and cluster signaling. When IL-6 binds to its membrane or soluble receptors, the co-receptor gp130 is activated to initiate downstream signaling and induce the expression of target genes. In the liver, IL-6 can perform its anti-inflammatory activities to promote hepatocyte reprogramming and liver regeneration. On the contrary, IL-6 also exerts the pro-inflammatory functions to induce liver aging, fibrosis, steatosis, and carcinogenesis. However, understanding the roles and underlying mechanisms of IL-6 in liver physiological and pathological processes is still an ongoing process. So far, therapeutic agents against IL‑6, IL‑6 receptor (IL‑6R), IL-6-sIL-6R complex, or IL-6 downstream signal transducers have been developed, and determined to be effective in the intervention of inflammatory diseases and cancers. In this review, we summarized and highlighted the understanding of the double-edged effects of IL-6 in liver homeostasis, aging, inflammation, and chronic diseases, for better shifting the "negative" functions of IL-6 to the "beneficial" actions, and further discussed the potential therapeutic effects of targeting IL-6 signaling in the clinics.
Collapse
Affiliation(s)
- Min-Jun Wang
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University (Naval Medical University), Shanghai, China.
| | - Hai-Ling Zhang
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University (Naval Medical University), Shanghai, China
- Department of Neurology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Fei Chen
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Xiao-Jing Guo
- Department of Health Statistics, Faculty of Health Service, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Qing-Gui Liu
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Jin Hou
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University (Naval Medical University), Shanghai, China.
| |
Collapse
|
25
|
Liu G, Zhang L, Zhou X, Xue J, Xia R, Gan X, Lv C, Zhang Y, Mao X, Kou X, Shi S, Chen Z. Inducing the "re-development state" of periodontal ligament cells via tuning macrophage mediated immune microenvironment. J Adv Res 2024; 60:233-248. [PMID: 37597747 PMCID: PMC11156709 DOI: 10.1016/j.jare.2023.08.009] [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: 04/18/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023] Open
Abstract
INTRODUCTION Periodontal regeneration, specifically the restoration of the cementum-periodontal ligament (PDL)-alveolar bone complex, remains a formidable challenge in the field of regenerative dentistry. In light of periodontal development, harnessing the multi-tissue developmental capabilities of periodontal ligament cells (PDLCs) and reinitiating the periodontal developmental process hold great promise as an effective strategy to foster the regeneration of the periodontal complex. OBJECTIVES This study aims to delve into the potential effects of the macrophage-mediated immune microenvironment on the "developmental engineering" regeneration strategy and its underlying molecular mechanisms. METHODS In this study, we conducted a comprehensive examination of the periodontium developmental process in the rat mandibular first molar using histological staining. Through the induction of diverse immune microenvironments in macrophages, we evaluated their potential effects on periodontal re-development events using a cytokine array. Additionally, we investigated PDLC-mediated periodontal re-development events under these distinct immune microenvironments through transcriptome sequencing and relevant functional assays. Furthermore, the underlying molecular mechanism was also performed. RESULTS The activation of development-related functions in PDLCs proved challenging due to their declined activity. However, our findings suggest that modulating the macrophage immune response can effectively regulate PDLCs-mediated periodontium development-related events. The M1 type macrophage immune microenvironment was found to promote PDLC activities associated with epithelial-mesenchymal transition, fiber degradation, osteoclastogenesis, and inflammation through the Wnt, IL-17, and TNF signaling pathways. Conversely, the M2 type macrophage immune microenvironment demonstrated superiority in inducing epithelium induction, fibers formation, and mineralization performance of PDLCs by upregulating the TGFβ and PI3K-Akt signaling pathway. CONCLUSION The results of this study could provide some favorable theoretical bases for applying periodontal development engineering strategy in resolving the difficulties in periodontal multi-tissue regeneration.
Collapse
Affiliation(s)
- Guanqi Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Linjun Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Xuan Zhou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Junlong Xue
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Ruidi Xia
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Xuejing Gan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Chunxiao Lv
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Yanshu Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Xueli Mao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; South China Center of Craniofacial Stem Cell Research,510055, Guangzhou, China
| | - Xiaoxing Kou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; South China Center of Craniofacial Stem Cell Research,510055, Guangzhou, China
| | - Songtao Shi
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; South China Center of Craniofacial Stem Cell Research,510055, Guangzhou, China
| | - Zetao Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China.
| |
Collapse
|
26
|
Xu J, Guo P, Hao S, Shangguan S, Shi Q, Volpe G, Huang K, Zuo J, An J, Yuan Y, Cheng M, Deng Q, Zhang X, Lai G, Nan H, Wu B, Shentu X, Wu L, Wei X, Jiang Y, Huang X, Pan F, Song Y, Li R, Wang Z, Liu C, Liu S, Li Y, Yang T, Xu Z, Du W, Li L, Ahmed T, You K, Dai Z, Li L, Qin B, Li Y, Lai L, Qin D, Chen J, Fan R, Li Y, Hou J, Ott M, Sharma AD, Cantz T, Schambach A, Kristiansen K, Hutchins AP, Göttgens B, Maxwell PH, Hui L, Xu X, Liu L, Chen A, Lai Y, Esteban MA. A spatiotemporal atlas of mouse liver homeostasis and regeneration. Nat Genet 2024; 56:953-969. [PMID: 38627598 DOI: 10.1038/s41588-024-01709-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/06/2024] [Indexed: 05/09/2024]
Abstract
The mechanism by which mammalian liver cell responses are coordinated during tissue homeostasis and perturbation is poorly understood, representing a major obstacle in our understanding of many diseases. This knowledge gap is caused by the difficulty involved with studying multiple cell types in different states and locations, particularly when these are transient. We have combined Stereo-seq (spatiotemporal enhanced resolution omics-sequencing) with single-cell transcriptomic profiling of 473,290 cells to generate a high-definition spatiotemporal atlas of mouse liver homeostasis and regeneration at the whole-lobe scale. Our integrative study dissects in detail the molecular gradients controlling liver cell function, systematically defining how gene networks are dynamically modulated through intercellular communication to promote regeneration. Among other important regulators, we identified the transcriptional cofactor TBL1XR1 as a rheostat linking inflammation to Wnt/β-catenin signaling for facilitating hepatocyte proliferation. Our data and analytical pipelines lay the foundation for future high-definition tissue-scale atlases of organ physiology and malfunction.
Collapse
Affiliation(s)
- Jiangshan Xu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Pengcheng Guo
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China.
- 3DC STAR, Spatiotemporal Campus at BGI Shenzhen, Shenzhen, China.
| | - Shijie Hao
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shuncheng Shangguan
- BGI Research, Shenzhen, China
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health and Guangzhou Medical University, Guangzhou, China
| | - Quan Shi
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Giacomo Volpe
- Hematology and Cell Therapy Unit, IRCCS-Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Keke Huang
- Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jing Zuo
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Juan An
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yue Yuan
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Mengnan Cheng
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Qiuting Deng
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiao Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Guangyao Lai
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health and Guangzhou Medical University, Guangzhou, China
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Haitao Nan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Baihua Wu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xinyi Shentu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Liang Wu
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaoyu Wei
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Yujia Jiang
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Xin Huang
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Fengyu Pan
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Yumo Song
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Ronghai Li
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Zhifeng Wang
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Chuanyu Liu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China
| | - Shiping Liu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | | | - Tao Yang
- China National GeneBank, BGI Research, Shenzhen, China
- Guangdong Provincial Genomics Data Center, BGI Research, Shenzhen, China
| | - Zhicheng Xu
- China National GeneBank, BGI Research, Shenzhen, China
- Guangdong Provincial Genomics Data Center, BGI Research, Shenzhen, China
| | - Wensi Du
- China National GeneBank, BGI Research, Shenzhen, China
- Guangdong Provincial Genomics Data Center, BGI Research, Shenzhen, China
| | - Ling Li
- China National GeneBank, BGI Research, Shenzhen, China
- Guangdong Provincial Genomics Data Center, BGI Research, Shenzhen, China
| | - Tanveer Ahmed
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Kai You
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhen Dai
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Li Li
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Baoming Qin
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yinxiong Li
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Liangxue Lai
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Dajiang Qin
- The Fifth Affiliated Hospital of Guangzhou Medical University-BGI Research Center for Integrative Biology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Junling Chen
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, China
| | - Rong Fan
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, China
| | - Yongyin Li
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, China
| | - Jinlin Hou
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, China
| | - Michael Ott
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Amar Deep Sharma
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Tobias Cantz
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | | - Andrew P Hutchins
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Berthold Göttgens
- Department of Haematology and Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Patrick H Maxwell
- Cambridge Institute for Medical Research, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lijian Hui
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xun Xu
- BGI Research, Hangzhou, China.
- BGI Research, Shenzhen, China.
- BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, Shenzhen, China.
| | - Longqi Liu
- BGI Research, Hangzhou, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
- BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China.
| | - Ao Chen
- BGI Research, Shenzhen, China.
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
- BGI Research, Chongqing, China.
- JFL-BGI STOmics Center, BGI-Shenzhen, Chongqing, China.
| | - Yiwei Lai
- BGI Research, Hangzhou, China.
- BGI Research, Shenzhen, China.
- 3DC STAR, Spatiotemporal Campus at BGI Shenzhen, Shenzhen, China.
- BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China.
| | - Miguel A Esteban
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China.
- 3DC STAR, Spatiotemporal Campus at BGI Shenzhen, Shenzhen, China.
- Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
- The Fifth Affiliated Hospital of Guangzhou Medical University-BGI Research Center for Integrative Biology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
27
|
Wu B, Shentu X, Nan H, Guo P, Hao S, Xu J, Shangguan S, Cui L, Cen J, Deng Q, Wu Y, Liu C, Song Y, Lin X, Wang Z, Yuan Y, Ma W, Li R, Li Y, Qian Q, Du W, Lai T, Yang T, Liu C, Ma X, Chen A, Xu X, Lai Y, Liu L, Esteban MA, Hui L. A spatiotemporal atlas of cholestatic injury and repair in mice. Nat Genet 2024; 56:938-952. [PMID: 38627596 DOI: 10.1038/s41588-024-01687-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 02/09/2024] [Indexed: 05/09/2024]
Abstract
Cholestatic liver injuries, characterized by regional damage around the bile ductular region, lack curative therapies and cause considerable mortality. Here we generated a high-definition spatiotemporal atlas of gene expression during cholestatic injury and repair in mice by integrating spatial enhanced resolution omics sequencing and single-cell transcriptomics. Spatiotemporal analyses revealed a key role of cholangiocyte-driven signaling correlating with the periportal damage-repair response. Cholangiocytes express genes related to recruitment and differentiation of lipid-associated macrophages, which generate feedback signals enhancing ductular reaction. Moreover, cholangiocytes express high TGFβ in association with the conversion of liver progenitor-like cells into cholangiocytes during injury and the dampened proliferation of periportal hepatocytes during recovery. Notably, Atoh8 restricts hepatocyte proliferation during 3,5-diethoxycarbonyl-1,4-dihydro-collidin damage and is quickly downregulated after injury withdrawal, allowing hepatocytes to respond to growth signals. Our findings lay a keystone for in-depth studies of cellular dynamics and molecular mechanisms of cholestatic injuries, which may further develop into therapies for cholangiopathies.
Collapse
Affiliation(s)
- Baihua Wu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xinyi Shentu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Haitao Nan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | - Shijie Hao
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jiangshan Xu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Shuncheng Shangguan
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health and Guangzhou Medical University, Guangzhou, China
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- BGI Research, Shenzhen, China
| | - Lei Cui
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jin Cen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiuting Deng
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Yan Wu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Chang Liu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Yumo Song
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Xiumei Lin
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | | | - Yue Yuan
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Wen Ma
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Ronghai Li
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Yikang Li
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Qiwei Qian
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Wensi Du
- China National GeneBank, BGI Research, Shenzhen, China
| | - Tingting Lai
- China National GeneBank, BGI Research, Shenzhen, China
| | - Tao Yang
- China National GeneBank, BGI Research, Shenzhen, China
| | - Chuanyu Liu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China
| | - Xiong Ma
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Ao Chen
- BGI Research, Shenzhen, China
| | - Xun Xu
- BGI Research, Shenzhen, China
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China
| | - Yiwei Lai
- BGI Research, Hangzhou, China.
- BGI Research, Shenzhen, China.
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China.
| | - Longqi Liu
- BGI Research, Hangzhou, China.
- BGI Research, Shenzhen, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
- China National GeneBank, BGI Research, Shenzhen, China.
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China.
| | - Miguel A Esteban
- BGI Research, Hangzhou, China.
- BGI Research, Shenzhen, China.
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
- The Fifth Affiliated Hospital of Guangzhou Medical University-BGI Research Center for Integrative Biology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Lijian Hui
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
28
|
Antonucci L, Karin M. The Past and Future of Inflammation as a Target to Cancer Prevention. Cancer Prev Res (Phila) 2024; 17:141-155. [PMID: 38271694 PMCID: PMC10987280 DOI: 10.1158/1940-6207.capr-23-0423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/21/2023] [Accepted: 01/23/2024] [Indexed: 01/27/2024]
Abstract
Inflammation is an essential defense mechanism in which innate immune cells are coordinately activated on encounter of harmful stimuli, including pathogens, tissue injury, and toxic compounds and metabolites to neutralize and eliminate the instigator and initiate healing and regeneration. Properly terminated inflammation is vital to health, but uncontrolled runaway inflammation that becomes chronic begets a variety of inflammatory and metabolic diseases and increases cancer risk. Making damaged tissues behave as "wounds that do not heal" and sustaining the production of growth factors whose physiologic function is tissue healing, chronic inflammation accelerates cancer emergence from premalignant lesions. In 1863, Rudolf Virchow, a leading German pathologist, suggested a possible association between inflammation and tumor formation, but it took another 140 years to fully elucidate and appreciate the tumorigenic role of inflammation. Key findings outlined molecular events in the inflammatory cascade that promote cancer onset and progression and enabled a better appreciation of when and where inflammation should be inhibited. These efforts triggered ongoing research work to discover and develop inflammation-reducing chemopreventive strategies for decreasing cancer risk and incidence.
Collapse
Affiliation(s)
- Laura Antonucci
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego School of Medicine; La Jolla, CA 92093, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego School of Medicine; La Jolla, CA 92093, USA
| |
Collapse
|
29
|
Liu Y, Xiao J, Cai J, Li R, Sui X, Zhang J, Lu T, Chen H, Chen G, Li H, Jiang C, Zhao X, Xiao C, Lei Y, Yao J, Lv G, Liang J, Zhang Y, Yang JR, Zheng J, Yang Y. Single-cell immune profiling of mouse liver aging reveals Cxcl2+ macrophages recruit neutrophils to aggravate liver injury. Hepatology 2024; 79:589-605. [PMID: 37695548 PMCID: PMC10871588 DOI: 10.1097/hep.0000000000000590] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 07/21/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND AND AIMS Immune cells play a crucial role in liver aging. However, the impact of dynamic changes in the local immune microenvironment on age-related liver injury remains poorly understood. We aimed to characterize intrahepatic immune cells at different ages to investigate key mechanisms associated with liver aging. APPROACH AND RESULTS We carried out single-cell RNA sequencing on mouse liver tissues at 4 different ages, namely, the newborn, suckling, young, and aged stages. The transcriptomic landscape, cellular classification, and intercellular communication were analyzed. We confirmed the findings by multiplex immunofluorescence staining, flow cytometry, in vitro functional experiments, and chimeric animal models. Nine subsets of 89,542 immune cells with unique properties were identified, of which Cxcl2+ macrophages within the monocyte/macrophage subset were preferentially enriched in the aged liver. Cxcl2+ macrophages presented a senescence-associated secretory phenotype and recruited neutrophils to the aged liver through the CXCL2-CXCR2 axis. Through the secretion of IL-1β and TNF-α, Cxcl2+ macrophages stimulated neutrophil extracellular traps formation. Targeting the CXCL2-CXCR2 axis limited the neutrophils migration toward the liver and attenuated age-related liver injury. Moreover, the relationship between Cxcl2+ macrophages and neutrophils in age-related liver injury was further validated by human liver transplantation samples. CONCLUSIONS This in-depth study illustrates that the mechanism of Cxcl2+ macrophage-driven neutrophil activation involves the CXCL2-CXCR2 axis and provides a potential therapeutic strategy for age-related liver injury.
Collapse
Affiliation(s)
- Yasong Liu
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Jiaqi Xiao
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Jianye Cai
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Rong Li
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Xin Sui
- Surgical ICU, The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Jiebin Zhang
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Tongyu Lu
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Haitian Chen
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Guihua Chen
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Haibo Li
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Chenhao Jiang
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Xuegang Zhao
- Surgical ICU, The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Cuicui Xiao
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University; Guangzhou, China
| | - Yunguo Lei
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Jia Yao
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Guo Lv
- Biological Treatment Center, The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Jinliang Liang
- Organ Transplantation Research Center of Guangdong Province Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yingcai Zhang
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Jian-Rong Yang
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-sen University; Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University; Guangzhou, China
| | - Jun Zheng
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Yang Yang
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| |
Collapse
|
30
|
Wang C, Bai Y, Li T, Liu J, Wang Y, Ju S, Yao W, Xiong B. Ginkgetin exhibits antifibrotic effects by inducing hepatic stellate cell apoptosis via STAT1 activation. Phytother Res 2024; 38:1367-1380. [PMID: 38217097 DOI: 10.1002/ptr.8106] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/12/2023] [Accepted: 12/16/2023] [Indexed: 01/15/2024]
Abstract
Liver fibrosis affects approximately 800 million patients worldwide, with over 2 million deaths each year. Nevertheless, there are no approved medications for treating liver fibrosis. In this study, we investigated the impacts of ginkgetin on liver fibrosis and the underlying mechanisms. The impacts of ginkgetin on liver fibrosis were assessed in mouse models induced by thioacetamide or bile duct ligation. Experiments on human LX-2 cells and primary mouse hepatic stellate cells (HSCs) were performed to explore the underlying mechanisms, which were also validated in the mouse models. Ginkgetin significantly decreased hepatic extracellular matrix deposition and HSC activation in the fibrotic models induced by thioacetamide (TAA) and bile duct ligation (BDL). Beneficial effects also existed in inhibiting hepatic inflammation and improving liver function. In vitro experiments showed that ginkgetin markedly inhibited HSC viability and induced HSC apoptosis dose-dependently. Mechanistic studies revealed that the antifibrotic effects of ginkgetin depend on STAT1 activation, as the effects were abolished in vitro after STAT1 silencing and in vivo after inhibiting STAT1 activation by fludarabine. Moreover, we observed a meaningful cross-talk between HSCs and hepatocytes, in which IL-6, released by ginkgetin-induced apoptotic HSCs, enhanced hepatocyte proliferation by activating STAT3 signaling. Ginkgetin exhibits antifibrotic effects by inducing HSC apoptosis via STAT1 activation and enhances hepatocyte proliferation secondary to HSC apoptosis via the IL-6/STAT3 pathway.
Collapse
Affiliation(s)
- Chaoyang Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaowei Bai
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tongqiang Li
- Department of Interventional Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiacheng Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingliang Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuguang Ju
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Xiong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Interventional Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
31
|
Katsuda T, Sussman JH, Ito K, Katznelson A, Yuan S, Takenaka N, Li J, Merrell AJ, Cure H, Li Q, Rasool RU, Asangani IA, Zaret KS, Stanger BZ. Cellular reprogramming in vivo initiated by SOX4 pioneer factor activity. Nat Commun 2024; 15:1761. [PMID: 38409161 PMCID: PMC10897393 DOI: 10.1038/s41467-024-45939-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 02/08/2024] [Indexed: 02/28/2024] Open
Abstract
Tissue damage elicits cell fate switching through a process called metaplasia, but how the starting cell fate is silenced and the new cell fate is activated has not been investigated in animals. In cell culture, pioneer transcription factors mediate "reprogramming" by opening new chromatin sites for expression that can attract transcription factors from the starting cell's enhancers. Here we report that SOX4 is sufficient to initiate hepatobiliary metaplasia in the adult mouse liver, closely mimicking metaplasia initiated by toxic damage to the liver. In lineage-traced cells, we assessed the timing of SOX4-mediated opening of enhancer chromatin versus enhancer decommissioning. Initially, SOX4 directly binds to and closes hepatocyte regulatory sequences via an overlapping motif with HNF4A, a hepatocyte master regulatory transcription factor. Subsequently, SOX4 exerts pioneer factor activity to open biliary regulatory sequences. The results delineate a hierarchy by which gene networks become reprogrammed under physiological conditions, providing deeper insight into the basis for cell fate transitions in animals.
Collapse
Affiliation(s)
- Takeshi Katsuda
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Jonathan H Sussman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
- Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kenji Ito
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Katznelson
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Salina Yuan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Naomi Takenaka
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jinyang Li
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Allyson J Merrell
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Hector Cure
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Qinglan Li
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Reyaz Ur Rasool
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Irfan A Asangani
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Kenneth S Zaret
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- The Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA.
| | - Ben Z Stanger
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA.
- The Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
32
|
Chen Y, Meng L, Xu N, Chen H, Wei X, Lu D, Wang S, Xu X. Ten-eleven translocation-2-mediated macrophage activation promotes liver regeneration. Cell Commun Signal 2024; 22:95. [PMID: 38308318 PMCID: PMC10835877 DOI: 10.1186/s12964-023-01407-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 11/23/2023] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND The remarkable regenerative capacity of the liver enables recovery after radical Hepatocellular carcinoma (HCC) resection. After resection, macrophages secrete interleukin 6 and hepatocyte growth factors to promote liver regeneration. Ten-eleven translocation-2 (Tet2) DNA dioxygenase regulates pro-inflammatory factor secretion in macrophages. In this study, we explored the role of Tet2 in macrophages and its function independent of its enzymatic activity in liver regeneration. METHODS The model of liver regeneration after 70% partial hepatectomy (PHx) is a classic universal model for studying reparative processes in the liver. Mice were euthanized at 0, 24, and 48 h after PHx. Enzyme-linked immunosorbent assays, quantitative reverse transcription-polymerase chain reaction, western blotting, immunofluorescence analysis, and flow cytometry were performed to explore immune cell infiltration and liver regenerative capability. Molecular dynamics simulations were performed to study the interaction between Tet2 and signal transducer and activator of transcription 1 (Stat1). RESULTS Tet2 in macrophages negatively regulated liver regeneration in the partial hepatectomy mice model. Tet2 interacted with Stat1, inhibiting the expression of proinflammatory factors and suppressing liver regeneration. The Tet2 inhibitor attenuated the interaction between Stat1 and Tet2, enhanced Stat1 phosphorylation, and promoted hepatocyte proliferation. The proliferative function of the Tet2 inhibitor relied on macrophages and did not affect hepatocytes directly. CONCLUSION Our findings underscore that Tet2 in macrophages negatively regulates liver regeneration by interacting with Stat1. Targeting Tet2 in macrophages promotes liver regeneration and function after a hepatectomy, presenting a novel target to promote liver regeneration and function.
Collapse
Affiliation(s)
- Yiyuan Chen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Lijun Meng
- Zhejiang University School of Medicine, Hangzhou, 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Nan Xu
- Zhejiang University School of Medicine, Hangzhou, 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Huan Chen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xuyong Wei
- Zhejiang University School of Medicine, Hangzhou, 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Di Lu
- Zhejiang University School of Medicine, Hangzhou, 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Shuai Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China.
| | - Xiao Xu
- Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China.
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
33
|
Zhang C, Sun C, Zhao Y, Ye B, Yu G. Signaling pathways of liver regeneration: Biological mechanisms and implications. iScience 2024; 27:108683. [PMID: 38155779 PMCID: PMC10753089 DOI: 10.1016/j.isci.2023.108683] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2023] Open
Abstract
The liver possesses a unique regenerative ability to restore its original mass, in this regard, partial hepatectomy (PHx) and partial liver transplantation (PLTx) can be executed smoothly and safely, which has important implications for the treatment of liver disease. Liver regeneration (LR) can be the very complicated procedure that involves multiple cytokines and transcription factors that interact with each other to activate different signaling pathways. Activation of these pathways can drive the LR process, which can be divided into three stages, namely, the initiation, progression, and termination stages. Therefore, it is important to investigate the pathways involved in LR to elucidate the mechanism of LR. This study reviews the latest research on the key signaling pathways in the different stages of LR.
Collapse
Affiliation(s)
- Chunyan Zhang
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
| | - Caifang Sun
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
| | - Yabin Zhao
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
| | - Bingyu Ye
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
| | - GuoYing Yu
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
| |
Collapse
|
34
|
Liu C, Xu Y, Yang G, Tao Y, Chang J, Wang S, Cheung TH, Chen J, Zeng YA. Niche inflammatory signals control oscillating mammary regeneration and protect stem cells from cytotoxic stress. Cell Stem Cell 2024; 31:89-105.e6. [PMID: 38141612 DOI: 10.1016/j.stem.2023.11.012] [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: 04/16/2023] [Revised: 09/03/2023] [Accepted: 11/29/2023] [Indexed: 12/25/2023]
Abstract
Stem cells are known for their resilience and enhanced activity post-stress. The mammary gland undergoes frequent remodeling and is subjected to recurring stress during the estrus cycle, but it remains unclear how mammary stem cells (MaSCs) respond to the stress and contribute to regeneration. We discovered that cytotoxic stress-induced activation of CD11c+ ductal macrophages aids stem cell survival and prevents differentiation. These macrophages boost Procr+ MaSC activity through IL1β-IL1R1-NF-κB signaling during the estrus cycle in an oscillating manner. Deleting IL1R1 in MaSCs results in stem cell loss and skewed luminal differentiation. Moreover, under cytotoxic stress from the chemotherapy agent paclitaxel, ductal macrophages secrete higher IL1β levels, promoting MaSC survival and preventing differentiation. Inhibiting IL1R1 sensitizes MaSCs to paclitaxel. Our findings reveal a recurring inflammatory process that regulates regeneration, providing insights into stress-induced inflammation and its impact on stem cell survival, potentially affecting cancer therapy efficacy.
Collapse
Affiliation(s)
- Chunye Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yishu Xu
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Guowei Yang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yu Tao
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiali Chang
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Shihui Wang
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research Center for Systems Biology and Human Health, the State Key Laboratory of Molecular Neuroscience, and Molecular Neuroscience Center, Hong Kong University of Science and Technology, Kowloon, Hong Kong, China; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518057, Guangdong, China
| | - Jianfeng Chen
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yi Arial Zeng
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
35
|
Cipriano A, Moqri M, Maybury-Lewis SY, Rogers-Hammond R, de Jong TA, Parker A, Rasouli S, Schöler HR, Sinclair DA, Sebastiano V. Mechanisms, pathways and strategies for rejuvenation through epigenetic reprogramming. NATURE AGING 2024; 4:14-26. [PMID: 38102454 PMCID: PMC11058000 DOI: 10.1038/s43587-023-00539-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 11/07/2023] [Indexed: 12/17/2023]
Abstract
Over the past decade, there has been a dramatic increase in efforts to ameliorate aging and the diseases it causes, with transient expression of nuclear reprogramming factors recently emerging as an intriguing approach. Expression of these factors, either systemically or in a tissue-specific manner, has been shown to combat age-related deterioration in mouse and human model systems at the cellular, tissue and organismal level. Here we discuss the current state of epigenetic rejuvenation strategies via partial reprogramming in both mouse and human models. For each classical reprogramming factor, we provide a brief description of its contribution to reprogramming and discuss additional factors or chemical strategies. We discuss what is known regarding chromatin remodeling and the molecular dynamics underlying rejuvenation, and, finally, we consider strategies to improve the practical uses of epigenetic reprogramming to treat aging and age-related diseases, focusing on the open questions and remaining challenges in this emerging field.
Collapse
Affiliation(s)
- Andrea Cipriano
- Department of Obstetrics & Gynecology, Stanford School of Medicine, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - Mahdi Moqri
- Department of Obstetrics & Gynecology, Stanford School of Medicine, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford School of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Tineke Anna de Jong
- Department of Obstetrics & Gynecology, Stanford School of Medicine, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - Alexander Parker
- Department of Obstetrics & Gynecology, Stanford School of Medicine, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - Sajede Rasouli
- Department of Obstetrics & Gynecology, Stanford School of Medicine, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - Hans Robert Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - David A Sinclair
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Paul F. Glenn Center for Biology of Aging Research, Harvard Medical School, Boston, MA, USA.
| | - Vittorio Sebastiano
- Department of Obstetrics & Gynecology, Stanford School of Medicine, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, USA.
| |
Collapse
|
36
|
Hu Y, Wang R, An N, Li C, Wang Q, Cao Y, Li C, Liu J, Wang Y. Unveiling the power of microenvironment in liver regeneration: an in-depth overview. Front Genet 2023; 14:1332190. [PMID: 38152656 PMCID: PMC10751322 DOI: 10.3389/fgene.2023.1332190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 11/29/2023] [Indexed: 12/29/2023] Open
Abstract
The liver serves as a vital regulatory hub for various physiological processes, including sugar, protein, and fat metabolism, coagulation regulation, immune system maintenance, hormone inactivation, urea metabolism, and water-electrolyte acid-base balance control. These functions rely on coordinated communication among different liver cell types, particularly within the liver's fundamental hepatic lobular structure. In the early stages of liver development, diverse liver cells differentiate from stem cells in a carefully orchestrated manner. Despite its susceptibility to damage, the liver possesses a remarkable regenerative capacity, with the hepatic lobule serving as a secure environment for cell division and proliferation during liver regeneration. This regenerative process depends on a complex microenvironment, involving liver resident cells, circulating cells, secreted cytokines, extracellular matrix, and biological forces. While hepatocytes proliferate under varying injury conditions, their sources may vary. It is well-established that hepatocytes with regenerative potential are distributed throughout the hepatic lobules. However, a comprehensive spatiotemporal model of liver regeneration remains elusive, despite recent advancements in genomics, lineage tracing, and microscopic imaging. This review summarizes the spatial distribution of cell gene expression within the regenerative microenvironment and its impact on liver regeneration patterns. It offers valuable insights into understanding the complex process of liver regeneration.
Collapse
Affiliation(s)
- Yuelei Hu
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun, China
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Ruilin Wang
- Department of Cadre’s Wards Ultrasound Diagnostics, Ultrasound Diagnostic Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Ni An
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Chen Li
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- College of Life Science and Bioengineering, Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, China
| | - Qi Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun, China
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Yannan Cao
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun, China
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Chao Li
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Juan Liu
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Yunfang Wang
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| |
Collapse
|
37
|
Tzouanas CN, Sherman MS, Shay JE, Rubin AJ, Mead BE, Dao TT, Butzlaff T, Mana MD, Kolb KE, Walesky C, Pepe-Mooney BJ, Smith CJ, Prakadan SM, Ramseier ML, Tong EY, Joung J, Chi F, McMahon-Skates T, Winston CL, Jeong WJ, Aney KJ, Chen E, Nissim S, Zhang F, Deshpande V, Lauer GM, Yilmaz ÖH, Goessling W, Shalek AK. Chronic metabolic stress drives developmental programs and loss of tissue functions in non-transformed liver that mirror tumor states and stratify survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.30.569407. [PMID: 38077056 PMCID: PMC10705501 DOI: 10.1101/2023.11.30.569407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Under chronic stress, cells must balance competing demands between cellular survival and tissue function. In metabolic dysfunction-associated steatotic liver disease (MASLD, formerly NAFLD/NASH), hepatocytes cooperate with structural and immune cells to perform crucial metabolic, synthetic, and detoxification functions despite nutrient imbalances. While prior work has emphasized stress-induced drivers of cell death, the dynamic adaptations of surviving cells and their functional repercussions remain unclear. Namely, we do not know which pathways and programs define cellular responses, what regulatory factors mediate (mal)adaptations, and how this aberrant activity connects to tissue-scale dysfunction and long-term disease outcomes. Here, by applying longitudinal single-cell multi -omics to a mouse model of chronic metabolic stress and extending to human cohorts, we show that stress drives survival-linked tradeoffs and metabolic rewiring, manifesting as shifts towards development-associated states in non-transformed hepatocytes with accompanying decreases in their professional functionality. Diet-induced adaptations occur significantly prior to tumorigenesis but parallel tumorigenesis-induced phenotypes and predict worsened human cancer survival. Through the development of a multi -omic computational gene regulatory inference framework and human in vitro and mouse in vivo genetic perturbations, we validate transcriptional (RELB, SOX4) and metabolic (HMGCS2) mediators that co-regulate and couple the balance between developmental state and hepatocyte functional identity programming. Our work defines cellular features of liver adaptation to chronic stress as well as their links to long-term disease outcomes and cancer hallmarks, unifying diverse axes of cellular dysfunction around core causal mechanisms.
Collapse
Affiliation(s)
- Constantine N. Tzouanas
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- These authors contributed equally
| | - Marc S. Sherman
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts, USA
- These authors contributed equally
| | - Jessica E.S. Shay
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Alcohol Liver Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- These authors contributed equally
| | - Adam J. Rubin
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Benjamin E. Mead
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tyler T. Dao
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Titus Butzlaff
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Miyeko D. Mana
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Kellie E. Kolb
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Chad Walesky
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian J. Pepe-Mooney
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Colton J. Smith
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sanjay M. Prakadan
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michelle L. Ramseier
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Evelyn Y. Tong
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Julia Joung
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, MA, Cambridge, MA, USA
- McGovern Institute for Brain Research at MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, MIT, Cambridge, MA, USA
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Fangtao Chi
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Thomas McMahon-Skates
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Carolyn L. Winston
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Woo-Jeong Jeong
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Katherine J. Aney
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ethan Chen
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sahar Nissim
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
- Gastroenterology Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Feng Zhang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, MA, Cambridge, MA, USA
- McGovern Institute for Brain Research at MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, MIT, Cambridge, MA, USA
| | - Vikram Deshpande
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Georg M. Lauer
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ömer H. Yilmaz
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA
- These senior authors contributed equally
| | - Wolfram Goessling
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA, USA
- These senior authors contributed equally
| | - Alex K. Shalek
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- These senior authors contributed equally
| |
Collapse
|
38
|
Jiang M, Ren J, Belmonte JCI, Liu GH. Hepatocyte reprogramming in liver regeneration: Biological mechanisms and applications. FEBS J 2023; 290:5674-5688. [PMID: 37556833 DOI: 10.1111/febs.16930] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/17/2023] [Accepted: 08/08/2023] [Indexed: 08/11/2023]
Abstract
The liver is one of the few organs that retain the capability to regenerate in adult mammals. This regeneration process is mainly facilitated by the dynamic behavior of hepatocytes, which are the major functional constituents in the liver. In response to liver injury, hepatocytes undergo remarkable alterations, such as reprogramming, wherein they lose their original identity and acquire properties from other cells. This phenomenon of hepatocyte reprogramming, coupled with hepatocyte expansion, plays a central role in liver regeneration, and its underlying mechanisms are complex and multifaceted. Understanding the fate of reprogrammed hepatocytes and the mechanisms of their conversion has significant implications for the development of innovative therapeutics for liver diseases. Herein, we review the plasticity of hepatocytes in response to various forms of liver injury, with a focus on injury-induced hepatocyte reprogramming. We provide a comprehensive summary of current knowledge on the molecular and cellular mechanisms governing hepatocyte reprogramming, specifically in the context of liver regeneration, providing insight into potential applications of this process in the treatment of liver disorders, including chronic liver diseases and liver cancer.
Collapse
Affiliation(s)
- Mengmeng Jiang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jie Ren
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of RNA Science and Engineering, CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
- Aging Biomarker Consortium, Beijing, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, China
| | | | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Aging Biomarker Consortium, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
39
|
Maowulieti G, Zhao S, Zhao M, Yuan H. The role of inflammatory factors and T-cell subsets in the diagnosis of recurrence in epithelial ovarian cancer patients and the effect of olaparin treatment on them. Immun Inflamm Dis 2023; 11:e1059. [PMID: 37904699 PMCID: PMC10599274 DOI: 10.1002/iid3.1059] [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: 03/29/2023] [Revised: 09/18/2023] [Accepted: 10/11/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND The aim of the study is to investigate the role of serum inflammatory factors and T-cell subsets in the diagnosis of recurrence in epithelial ovarian cancer patients and the effect of olaparib on inflammatory factor and T-lymphocyte subsets in patients with recurrent epithelial ovarian cancer. METHODS In this study, 100 patients diagnosed as recurrent epithelial ovarian cancer in our hospital and 100 patients without recurrent epithelial ovarian cancer in the same period were selected. According to the treatment plan, the recurrent patients were divided into conventional therapy group (Paclitaxel and Carboplatin) and combined therapy group (Paclitaxel, Carboplatin, and olaparib). The levels of serum inflammatory factors were evaluated by enzyme-linked immunosorbent assay. The peripheral blood T-lymphocyte subsets in each group were detected by flow cytometry. RESULTS Compared with nonrecurrent patients, recurrent patients have higher serum interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) levels (p < .05), and lower interferon-γ (IFN-γ) level and the CD4+/CD8+ ratio. After adjusting for confounding factors, the results showed that the serum IL-6, IFN-γ, and TNF-α levels were influencing factors of recurrence in epithelial ovarian cancer patients. The area under the receiver operating curve and the sensitivity of serum TNF-α in predicting ovarian cancer recurrence were higher than those of IL-6 and IFN-γ. After secondary chemotherapy and/or olaparib maintenance treatment, the IL-6 (p < .001) and TNF-α (p < .001) levels in combined therapy group were lower than those in the conventional therapy, whereas the IFN-γ level (p < .001), the CD4+ T-cell proportion (p = .0069) and the CD4+/CD8+ ratio (p = .0201) were higher than those in the conventional therapy. CONCLUSION The serum IL-6, TNF-α, and IFN-γ levels were closely related to the recurrence of ovarian cancer. Olaparib maintenance treatment can significantly decrease the IL-6 and TNF-α level, and increase IFN-γ level and the CD4+/CD8+ ratio in patients with recurrent ovarian cancer.
Collapse
Affiliation(s)
- Gulijianati Maowulieti
- Department of Gynaecology, Wuxi Maternity and Child Health Care HospitalWomen's Hospital of Jiangnan University, Jiangnan UniversityWuxiJiangsuChina
| | - Shaojie Zhao
- Department of Gynaecology, Wuxi Maternity and Child Health Care HospitalWomen's Hospital of Jiangnan University, Jiangnan UniversityWuxiJiangsuChina
| | - Min Zhao
- Department of Gynaecology, Wuxi Maternity and Child Health Care HospitalWomen's Hospital of Jiangnan University, Jiangnan UniversityWuxiJiangsuChina
| | - Hua Yuan
- Department of Gynaecology, Wuxi Maternity and Child Health Care HospitalWomen's Hospital of Jiangnan University, Jiangnan UniversityWuxiJiangsuChina
| |
Collapse
|
40
|
Nagai H, Nagai LAE, Tasaki S, Nakato R, Umetsu D, Kuranaga E, Miura M, Nakajima Y. Nutrient-driven dedifferentiation of enteroendocrine cells promotes adaptive intestinal growth in Drosophila. Dev Cell 2023; 58:1764-1781.e10. [PMID: 37689060 DOI: 10.1016/j.devcel.2023.08.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 05/05/2023] [Accepted: 08/16/2023] [Indexed: 09/11/2023]
Abstract
Post-developmental organ resizing improves organismal fitness under constantly changing nutrient environments. Although stem cell abundance is a fundamental determinant of adaptive resizing, our understanding of its underlying mechanisms remains primarily limited to the regulation of stem cell division. Here, we demonstrate that nutrient fluctuation induces dedifferentiation in the Drosophila adult midgut to drive adaptive intestinal growth. From lineage tracing and single-cell RNA sequencing, we identify a subpopulation of enteroendocrine (EE) cells that convert into functional intestinal stem cells (ISCs) in response to dietary glucose and amino acids by activating the JAK-STAT pathway. Genetic ablation of EE-derived ISCs severely impairs ISC expansion and midgut growth despite the retention of resident ISCs, and in silico modeling further indicates that EE dedifferentiation enables an efficient increase in the midgut cell number while maintaining epithelial cell composition. Our findings identify a physiologically induced dedifferentiation that ensures ISC expansion during adaptive organ growth in concert with nutrient conditions.
Collapse
Affiliation(s)
- Hiroki Nagai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan.
| | | | - Sohei Tasaki
- Graduate School of Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Ryuichiro Nakato
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Daiki Umetsu
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan; Graduate School of Science, Osaka University, Osaka 560-0043, Japan
| | - Erina Kuranaga
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yuichiro Nakajima
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan; Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan.
| |
Collapse
|
41
|
Cui D, Xu D, Yue S, Yan C, Liu W, Fu R, Ma W, Tang Y. Recent advances in the pharmacological applications and liver toxicity of triptolide. Chem Biol Interact 2023; 382:110651. [PMID: 37516378 DOI: 10.1016/j.cbi.2023.110651] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/23/2023] [Accepted: 07/27/2023] [Indexed: 07/31/2023]
Abstract
Triptolide is a predominant active component of Triptergium wilfordii Hook. F, which has been used for the treatment of cancers and autoimmune diseases such as rheumatoid arthritis, systemic lupus erythematosus and diabetic nephropathy. Therefore, triptolide and its derivates are considered to have promising prospects for development into drugs. However, the clinical application of triptolide is limited due to various organ toxicities, especially liver toxicity. The potential mechanism of triptolide-induced hepatotoxicity has attracted increasing attention. Over the past five years, studies have revealed that triptolide-induced liver toxicity is involved in metabolic imbalance, oxidative stress, inflammations, autophagy, apoptosis, and the regulation of cytochrome P450 (CYP450) enzymes, gut microbiota and immune cells. In this review, we summarize the pharmacological applications and hepatotoxicity mechanism of triptolide, which will provide solid theoretical evidence for further research of triptolide.
Collapse
Affiliation(s)
- Dongxiao Cui
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Dingqiao Xu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Shijun Yue
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Chaoqun Yan
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Wenjuan Liu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Ruijia Fu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Wenfu Ma
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Yuping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an, China.
| |
Collapse
|
42
|
Wen Y. The Role of Immune Cells in Liver Regeneration. LIVERS 2023; 3:383-396. [DOI: 10.3390/livers3030029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
The liver is the only organ that can regenerate and regain its original tissue-to-body weight ratio within a short period of time after tissue loss. Insufficient liver regeneration in patients after partial hepatectomy or liver transplantation with partial liver grafts often leads to post-hepatectomy liver failure or small-for-size syndrome, respectively. Enhancing liver regeneration after liver injury might improve outcomes and increase patient survival. Liver regeneration comprises hepatocyte proliferation, and hepatic progenitor cell expansion and differentiation into hepatocytes. The immune system is intensively involved in liver regeneration. The current review provides a comprehensive overview of the diverse roles played by immune cells in liver regeneration. Macrophages, neutrophils, eosinophils, basophils, mast cells, platelets, dendritic cells, type 1 innate lymphoid cells, B cells, and T cells are implicated in promoting liver regeneration, while natural killer cells and overactivated natural killer T cells are supposed to inhibit hepatocyte proliferation. We also highlight the predominant underlying mechanisms mediated by immune cells, which may contribute to the development of novel strategies for promoting liver regeneration in patients with liver diseases.
Collapse
Affiliation(s)
- Yankai Wen
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
43
|
Chen F, Schönberger K, Tchorz JS. Distinct hepatocyte identities in liver homeostasis and regeneration. JHEP Rep 2023; 5:100779. [PMID: 37456678 PMCID: PMC10339260 DOI: 10.1016/j.jhepr.2023.100779] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 07/18/2023] Open
Abstract
The process of metabolic liver zonation is spontaneously established by assigning distributed tasks to hepatocytes along the porto-central blood flow. Hepatocytes fulfil critical metabolic functions, while also maintaining hepatocyte mass by replication when needed. Recent technological advances have enabled us to fine-tune our understanding of hepatocyte identity during homeostasis and regeneration. Subsets of hepatocytes have been identified to be more regenerative and some have even been proposed to function like stem cells, challenging the long-standing view that all hepatocytes are similarly capable of regeneration. The latest data show that hepatocyte renewal during homeostasis and regeneration after liver injury is not limited to rare hepatocytes; however, hepatocytes are not exactly the same. Herein, we review the known differences that give individual hepatocytes distinct identities, recent findings demonstrating how these distinct identities correspond to differences in hepatocyte regenerative capacity, and how the plasticity of hepatocyte identity allows for division of labour among hepatocytes. We further discuss how these distinct hepatocyte identities may play a role during liver disease.
Collapse
Affiliation(s)
- Feng Chen
- Novartis Institutes for BioMedical Research, Cambridge, MA, United States
| | | | - Jan S. Tchorz
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| |
Collapse
|
44
|
Jin Y, Guo YH, Li JC, Li Q, Ye D, Zhang XX, Li JT. Vascular endothelial growth factor protein and gene delivery by novel nanomaterials for promoting liver regeneration after partial hepatectomy. World J Gastroenterol 2023; 29:3748-3757. [PMID: 37426320 PMCID: PMC10324527 DOI: 10.3748/wjg.v29.i24.3748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/13/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
Partial hepatectomy (PH) can lead to severe complications, including liver failure, due to the low regenerative capacity of the remaining liver, especially after extensive hepatectomy. Liver sinusoidal endothelial cells (LSECs), whose proliferation occurs more slowly and later than hepatocytes after PH, compose the lining of the hepatic sinusoids, which are the smallest blood vessels in the liver. Vascular endothelial growth factor (VEGF), secreted by hepatocytes, promotes LSEC proliferation. Supplementation of exogenous VEGF after hepatectomy also increases the number of LSECs in the remaining liver, thus promoting the reestablishment of the hepatic sinusoids and accelerating liver regeneration. At present, some shortcomings exist in the methods of supplementing exogenous VEGF, such as a low drug concentration in the liver and the reaching of other organs. More-over, VEGF should be administered multiple times and in large doses because of its short half-life. This review summarized the most recent findings on liver regeneration and new strategies for the localized delivery VEGF in the liver.
Collapse
Affiliation(s)
- Yun Jin
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang Province, China
| | - Ying-Hao Guo
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang Province, China
| | - Jia-Cheng Li
- Department of General Surgery, Yuhuan Second People’s Hospital, Taizhou 317600, Zhejiang Province, China
| | - Qi Li
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang Province, China
| | - Dan Ye
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang Province, China
| | - Xiao-Xiao Zhang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang Province, China
| | - Jiang-Tao Li
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang Province, China
| |
Collapse
|
45
|
Du Y, Jian S, Wang X, Yang C, Qiu H, Fang K, Yan Y, Shi J, Li J. Machine learning and single cell RNA sequencing analysis identifies regeneration-related hepatocytes and highlights a Birc5-related model for identifying cell proliferative ability. Aging (Albany NY) 2023; 15:204775. [PMID: 37315292 PMCID: PMC10292894 DOI: 10.18632/aging.204775] [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: 03/21/2023] [Accepted: 05/17/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Partial hepatectomy (PHx) has been shown to induce rapid regeneration of adult liver under emergency conditions. Therefore, an in-depth investigation of the underlying mechanisms that govern liver regeneration following PHx is crucial for a comprehensive understanding of this process. METHOD We analyzed scRNA-seq data from liver samples of normal and PHx-48-hour mice. Seven machine learning algorithms were utilized to screen and validate a gene signature that accurately identifies and predicts this population. Co-immunostaining of zonal markers with BIRC5 to investigate regional characteristics of hepatocytes post-PHx. RESULTS Single cell sequencing results revealed a population of regeneration-related hepatocytes. Transcription factor analysis emphasized the importance of Hmgb1 transcription factor in liver regeneration. HdWGCNA and machine learning algorithm screened and obtained the key signature characterizing this population, including a total of 17 genes and the function enrichment analysis indicated their high correlation with cell cycle pathway. It is note-worthy that we inferred that Hmgb1 might be vital in the regeneration-related hepatocytes of PHx_48h group. Parallelly, Birc5 might be closely related to the regulation of liver regeneration, and positively correlated with Hmgb1. CONCLUSIONS Our study has identified a distinct population of hepatocytes that are closely associated with liver regeneration. Through machine learning algorithms, we have identified a set of 17 genes that are highly indicative of the regenerative capacity of hepatocytes. This gene signature has enabled us to assess the proliferation ability of in vitro cultured hepatocytes using sequencing data alone.
Collapse
Affiliation(s)
- Yuan Du
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shuqin Jian
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xicheng Wang
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Chao Yang
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Hua Qiu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Kang Fang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yehong Yan
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jun Shi
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of General Surgery, Ji’an Hospital of Shanghai East Hospital, School of Medicine, Tongji University, Ji’an, Jiangxi, China
| | - Jianfeng Li
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
46
|
Ke C, Shu L, Cai L, Yujun Z, Qiang W. IGF2BP3/HIF1A/YAP signaling plays a role in driving acute-on-chronic liver failure through activating hepatocyte reprogramming. Cell Signal 2023:110727. [PMID: 37257765 DOI: 10.1016/j.cellsig.2023.110727] [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: 03/12/2023] [Revised: 05/03/2023] [Accepted: 05/18/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND Acute-on-chronic liver failure (ACLF) is a syndrome with both high prevalence and mortality. However, the underlying mechanisms remain elusive and there is no effective therapeutic approach available. Here we aim to uncover novel molecular mechanisms of ACLF and identify potential therapeutic targets. METHOD We performed integrative analysis of 3 transcriptomic datasets and subsequent bioinformatic analysis aiming for potential genes of significance in ACLF development, identifying a critical role of IGF2BP3/HIF1A signaling in development of ACLF. Expression of molecules in IGF2BP3/HIF1A pathway and hepatocyte reprogramming markers in clinical samples were then determined by western blot and quantitative PCR. N6-methyladenosine (m6A) RNA modification of HIF1A was analyzed by m6A dot assay and PCR following m6A-antibody precipitation. The molecular mechanisms among IGFBP3, HIF1α and YAP1 were further validated by gene overexpression and knockdown experiments in HepG2 and Hep3B cells. Cell phenotypes of hepatocyte reprogramming were determined by EdU staining, sphere formation assay and immunoblotting of relevant markers. RESULTS Our data demonstrated that IGF2BP3 recognized m6A modification in HIF1A mRNA as an m6A reader, thereby promoting expression of HIF1A by increasing RNA stability. HIF1A activated Rho GTPases (RhoA) and suppressed phosphorylation of YAP via inhibiting LATS1/2, promoting translocation of non-phosphorylated YAP into the nucleus, resulting in fetal liver programme and ultimate hepatic injury in ACLF patients. CONCLUSION We reveal a novel molecular mechanism that IGF2BP3/HIF1A/YAP signaling promotes hepatocyte reprogramming, causing hepatic injury in ACLF. Our study provides potential targets for treatment of ACLF.
Collapse
Affiliation(s)
- Cheng Ke
- Department of Transplantation, The Third Xiangya Hospital, Central South University, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, China
| | - Liu Shu
- Department of Transplantation, The Third Xiangya Hospital, Central South University, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, China
| | - Li Cai
- Department of Transplantation, The Third Xiangya Hospital, Central South University, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, China
| | - Zhao Yujun
- Department of Transplantation, The Third Xiangya Hospital, Central South University, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, China
| | - Wang Qiang
- Department of Transplantation, The Third Xiangya Hospital, Central South University, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, China.
| |
Collapse
|
47
|
VanHook AM. IL-6 drives hepatocyte dedifferentiation. Sci Signal 2023; 16:eadh4937. [PMID: 36917641 DOI: 10.1126/scisignal.adh4937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Injury-induced hepatocyte reprogramming depends on a proinflammatory cytokine from Kupffer cells.
Collapse
|