1
|
Cairoli V, Valle-Millares D, Ryan P, Dominguez L, Martín-Carbonero L, De los Santos I, De Matteo E, Ameigeiras B, De Sousa M, Briz V, Preciado MV, Fernández-Rodriguez A, Valva P. Extracellular vesicles derived microRNAs as non-invasive markers of liver fibrosis in chronically infected HCV patients: a pilot study. Noncoding RNA Res 2025; 12:132-140. [PMID: 40176849 PMCID: PMC11964596 DOI: 10.1016/j.ncrna.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 12/08/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Extracellular vesicles (EVs) are an increasingly promising tool for liquid biopsy in liver diseases. Hepatitis C Virus (HCV) infection, alone or together with Human Immunodeficiency Virus (HIV) infection significantly impacts on the microRNA (miRNA) EVs content resembling chronic hepatitis C (CHC) progression. The objective of the study was to delve into the intricate EVs-miRNA profiles in CHC patients with different liver fibrosis stages, aiming to pinpoint non-invasive markers capable of distinguishing significant fibrosis. Plasma EV-miRNAs from 50 CHC patients (HCV+ and HCV+/HIV+) stratified in no significant (F < 2) and significant (F ≥ 2) fibrosis, were massively sequenced. General linear models (GLM) were used to identify significantly differential expressed (SDE) miRNAs according to liver fibrosis stages (F ≥ 2 and F < 2). Dysregulated biological pathways were subsequently analyzed in silico for the following groups: i) all patients; ii) HCV+; and iii) HCV+/HIV+. Multiple-ordered logistic regression analysis was performed to develop a score to identify F ≥ 2 cases. The diagnostic potential of both the SDE miRNAs and the developed score was assessed using ROC curve analysis. With respect to all CHC patients, two SDE miRNAs (hsa-miR-122-5p and hsa-miR-92a-3p) were identified which regulate genes related to cytoskeleton organization. Regarding their diagnostic performance to discriminate F ≥ 2, both miRNAs individually demonstrated acceptable diagnostic values. However, their combined use in a new score enhanced their diagnostic performance (AUROC = 0.833). In the HCV+ subgroup, 8 SDE miRNAs (hsa-miR-122-5p, hsa-miR-320c, hsa-miR-3615, hsa-miR-320a-3p, hsa-miR-374b-5p, hsa-let-7a-3p, hsa-miR-199a-5p, hsa-miR-142-5p), which regulate macrophage activity and cell growth/death regulation, were recognized. Among them, hsa-miR-3615 displayed the highest diagnostic performance to discriminate F ≥ 2 (AUROC = 0.936). With respect to HCV+/HIV+, 18 SDE miRNAs (hsa-miR-4508, hsa-miR-122-5p, hsa-miR-451a, hsa-miR-1290, hsa-miR-1246, hsa-miR-107, hsa-miR-15b-5p, hsa-miR-194-5p, hsa-miR-22-5p, hsa-miR-20b-5p, hsa-miR-142-5p, hsa-miR-328-3p, hsa-miR-335-3p, hsa-miR-125a-5p, hsa-miR-423-3p, hsa-let-7d-3p, hsa-miR-128-3p, hsa-miR-10a-5p) were recognized that regulate RNA silencing processes. In this case, hsa-miR-423-3p and hsa-miR-128-3p showed outstanding diagnostic performances (AUROC > 0.900). Distinct EVs-miRNA profiles were identified in patients with varying liver fibrosis stages, both in the overall CHC cohort and within HCV+ and HCV+/HIV+ subgroups. These specific miRNA signatures would allow the elucidation of potential mechanisms involved in clinical evolution and identification of specific biomarkers of unfavorable progression, plausible to be used in a diagnostic panel. Furthermore, the developed score demonstrates the ability to discriminate within the CHC group those individuals with significant fibrosis regardless of their HIV infection status.
Collapse
Affiliation(s)
- Victoria Cairoli
- Multidisciplinary Institute for Investigation in Pediatric Pathologies (IMIPP), CONICET-GCBA, Laboratory of Molecular Biology, Pathology Division, Ricardo Gutiérrez Children's Hospital, C1425EFD CABA, Buenos Aires, Argentina
| | - Daniel Valle-Millares
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28222, Madrid, Spain
| | - Pablo Ryan
- Infectious Diseases Department, Internal Medicine Department HIV/Hepatitis, Infanta Leonor University Hospital, 28031, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28222, Madrid, Spain
| | - Lourdes Dominguez
- HIV Unit, Internal Medicine Department, Research Institute of the Hospital 12 de Octubre (imas12), 28041, Madrid, Spain
| | - Luz Martín-Carbonero
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28222, Madrid, Spain
- Infectious Diseases Unit, Internal Medicine Department, La Paz University Hospital, IdiPAZ, 28046, Madrid, Spain
| | - Ignacio De los Santos
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28222, Madrid, Spain
- Infectious Diseases Unit, Internal Medicine Department, La Princesa University Hospital, 28006, Madrid, Spain
| | - Elena De Matteo
- Multidisciplinary Institute for Investigation in Pediatric Pathologies (IMIPP), CONICET-GCBA, Laboratory of Molecular Biology, Pathology Division, Ricardo Gutiérrez Children's Hospital, C1425EFD CABA, Buenos Aires, Argentina
| | - Beatriz Ameigeiras
- Liver Unit, Ramos Mejía Hospital, C1221ADC CABA, Buenos Aires, Argentina
| | - Marcela De Sousa
- Liver Unit, Ramos Mejía Hospital, C1221ADC CABA, Buenos Aires, Argentina
| | - Verónica Briz
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28222, Madrid, Spain
- Viral Hepatitis Reference and Research Laboratory, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28222, Madrid, Spain
| | - María V. Preciado
- Multidisciplinary Institute for Investigation in Pediatric Pathologies (IMIPP), CONICET-GCBA, Laboratory of Molecular Biology, Pathology Division, Ricardo Gutiérrez Children's Hospital, C1425EFD CABA, Buenos Aires, Argentina
| | - Amanda Fernández-Rodriguez
- Unit of Viral Infection and Immunity, National Center for Microbiology (CNM), Health Institute Carlos III (ISCIII), Majadahonda, 28222, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28222, Madrid, Spain
| | - Pamela Valva
- Multidisciplinary Institute for Investigation in Pediatric Pathologies (IMIPP), CONICET-GCBA, Laboratory of Molecular Biology, Pathology Division, Ricardo Gutiérrez Children's Hospital, C1425EFD CABA, Buenos Aires, Argentina
| |
Collapse
|
2
|
Mukkala AN, David BA, Ailenberg M, Liang J, Vaswani CM, Karakas D, Goldfarb R, Barbour W, Gasner A, Wu RS, Petrut R, Jerkic M, Andreazza AC, Dos Santos C, Ni H, Zhang H, Kapus A, Kubes P, Rotstein OD. Mitochondrial Transplantation: A Novel Therapy for Liver Ischemia/Reperfusion Injury. Ann Surg 2025; 281:1032-1047. [PMID: 39912224 DOI: 10.1097/sla.0000000000006655] [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] [Indexed: 02/07/2025]
Abstract
OBJECTIVE To investigate the hepatoprotective effects of mitochondrial transplantation (MTx) in a murine liver ischemia/reperfusion (I/R) model. BACKGROUND Sequential liver ischemia, followed by reperfusion (I/R), is a pathophysiological process underlying hepatocellular injury in a number of clinical contexts, such as hemorrhagic shock/resuscitation, major elective liver surgery, and organ transplantation. A unifying pathogenic consequence of I/R is mitochondrial dysfunction. Restoration of mitochondria through transplantation (MTx) has emerged as a potential therapeutic in I/R. However, its role in liver I/R and its mechanisms of action remain poorly defined. METHODS We investigated the hepatoprotective effects of MTx in an in vivo mouse model of liver I/R and used in vivo imaging and various knockout and transgenic mouse models to determine the mechanism of protection. RESULTS We found that I/R-induced hepatocellular injury was prevented by MTx, as measured by plasma ALT, AST, and liver histology. In addition, I/R-induced pro-inflammatory cytokine release (IL-6, TNFα) was dampened by MTx, and anti-inflammatory IL-10 was enhanced. Moreover, MTx lowered neutrophil infiltration into both the liver sinusoids and lung bronchoalveolar lavage fluid, suggesting a local and distant reduction in inflammation. Using in vivo intravital imaging, we found that I/R-subjected Kupffer cells (KCs), rapidly sequestered transplanted mitochondria, and acidified mitochondria within lysosomal compartments. To specifically interrogate the role of KCs, we depleted KCs using the diphtheria toxin-inducible Clec4f/iDTR transgenic mouse, then induced I/R, and discovered that KCs are necessary for the beneficial effects of MTx. Finally, we induced I/R in the complement receptor of the immunoglobulin (CRIg) superfamily knockout mice and found that CRIg was required for mitochondria capture by KCs and mitochondria-mediated hepatoprotection. CONCLUSIONS In this study, we demonstrated that CRIg-dependent capture of mitochondria by I/R-subjected KCs is a hepatoprotective mechanism in vivo . These data progress knowledge on the mechanisms of MTx and open new avenues for clinical translation.
Collapse
Affiliation(s)
- Avinash Naraiah Mukkala
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto; Toronto, ON, Canada
| | - Bruna Araujo David
- Department of Physiology and Pharmacology, University of Calgary; Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary; Calgary, AB, Canada
| | - Menachem Ailenberg
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
| | - Jady Liang
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto; Toronto, ON, Canada
| | - Chirag Manoj Vaswani
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto; Toronto, ON, Canada
| | - Danielle Karakas
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto; Toronto, ON, Canada
| | - Rachel Goldfarb
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
| | - William Barbour
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
| | - Avishai Gasner
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
| | - Ruoxian Scarlet Wu
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
| | - Raluca Petrut
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
| | - Mirjana Jerkic
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
| | - Ana Cristina Andreazza
- Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto; Toronto, ON, Canada
| | - Claudia Dos Santos
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
- Interdepartmental Division of Critical Care, Temerty Faculty of Medicine, University of Toronto; Toronto, ON, Canada
| | - Heyu Ni
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto; Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto; Toronto, ON, Canada
- Canadian Blood Services Centre for Innovation, Canadian Blood Service; Toronto, ON, Canada
| | - Haibo Zhang
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto; Toronto, ON, Canada
- Interdepartmental Division of Critical Care, Temerty Faculty of Medicine, University of Toronto; Toronto, ON, Canada
- Department of Anesthesiology and Pain Medicine, Temerty Faculty of Medicine, University of Toronto; Toronto, ON, Canada
| | - Andras Kapus
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto; Toronto, ON, Canada
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary; Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary; Calgary, AB, Canada
| | - Ori David Rotstein
- Keenan Research Centre for Biomedical Science, Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto; Toronto, ON, Canada
- Department of Surgery, St. Michael's Hospital, Unity Health Toronto; Toronto, ON, Canada
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto; Toronto, ON, Canada
| |
Collapse
|
3
|
Chen Z, Zhang J, Zhang L, Liu Y, Zhang T, Sang X, Xu Y, Lu X. Identification of PANoptosis related biomarkers to predict hepatic ischemia‒reperfusion injury after liver transplantation. Sci Rep 2025; 15:15437. [PMID: 40316717 PMCID: PMC12048552 DOI: 10.1038/s41598-025-99264-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 04/18/2025] [Indexed: 05/04/2025] Open
Abstract
Hepatic ischemia-reperfusion injury (HIRI) is a major complication following liver transplantation. Bioinformatic analysis was performed to elucidate the PANoptosis-related molecular mechanisms underlying HIRI. Comprehensive analysis of bulk and single-cell RNA sequencing data from human liver tissue before and after HIRI was performed. Differential expression analysis, weighted gene coexpression analysis, and protein interaction network analysis were used to identify candidate biomarkers. Multiple machine learning methods were utilized to screen for core biomarkers and construct a diagnostic predictive model. Functional and interaction analyses of the genes were also performed. Cellular clustering and annotation, pseudotemporal trajectory, and intercellular communication analyses of HIRI were conducted. Six PANoptosis-associated genes (CEBPB, HSPA1A, HSPA1B, IRF1, SERPINE1, and TNFAIP3) were identified as HIRI-related biomarkers. These biomarkers are regulated by NF-κB and miRNA-155. A nomogram for HIRI prediction based on these biomarkers was constructed and validated. In addition, the heterogeneity and dynamic changes in macrophage subpopulations during HIRI were revealed, highlighting the roles of Kupffer cells and monocyte-derived macrophages in modulating the hepatic microenvironment. The MIF and VISFATIN signaling pathways play important roles in the interaction between macrophages and other cells. These findings enhance our understanding of the mechanisms of PANoptosis in HIRI and provide a new basis and potential targets for prevention and treatment strategies for HIRI.
Collapse
Affiliation(s)
- Zhihong Chen
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Junwei Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Lei Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Yaoge Liu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Ting Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Xinting Sang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Yiyao Xu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China.
| | - Xin Lu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
4
|
Obrzut O, Gostyńska-Stawna A, Kustrzyńska K, Stawny M, Krajka-Kuźniak V. Curcumin: A Natural Warrior Against Inflammatory Liver Diseases. Nutrients 2025; 17:1373. [PMID: 40284236 PMCID: PMC12030243 DOI: 10.3390/nu17081373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
Curcumin (CUR), a bioactive compound found in turmeric, has garnered attention for its potential anti-inflammatory properties and impact on liver health. Numerous studies suggest that CUR may be crucial in mitigating liver inflammation. The compound's anti-inflammatory effects are believed to be attributed to its ability to modulate various molecular pathways involved in the inflammatory response. Research indicates that CUR may suppress the activation of inflammatory cells and the production of pro-inflammatory cytokines in the liver. Additionally, it has been observed to inhibit the activity of transcription factors that play a key role in inflammation. By targeting these molecular mechanisms, CUR may help alleviate the inflammatory burden on the liver. Moreover, CUR's antioxidant properties are thought to contribute to its protective effects on the liver. Oxidative stress is closely linked to inflammation, and CUR's ability to neutralize free radicals may further support its anti-inflammatory action. While the evidence is promising, it is essential to note that more research is needed to fully understand the precise mechanisms through which CUR influences liver inflammation. Nevertheless, these findings suggest that CUR could be a potential therapeutic agent in managing liver inflammatory conditions. In this review, we explore the potential impact of CUR on inflammation, highlighting the key mechanisms involved, as reported in the literature.
Collapse
Affiliation(s)
- Olga Obrzut
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
| | - Aleksandra Gostyńska-Stawna
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (A.G.-S.); (M.S.)
| | - Karolina Kustrzyńska
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
- Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland
| | - Maciej Stawny
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (A.G.-S.); (M.S.)
| | - Violetta Krajka-Kuźniak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
| |
Collapse
|
5
|
Hong J, Kim YH. Cutting-edge biotherapeutics and advanced delivery strategies for the treatment of metabolic dysfunction-associated steatotic liver disease spectrum. J Control Release 2025; 380:433-456. [PMID: 39923856 DOI: 10.1016/j.jconrel.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/22/2024] [Accepted: 02/04/2025] [Indexed: 02/11/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), a condition with the potential to progress into liver cirrhosis or hepatocellular carcinoma, has become a significant global health concern due to its increasing prevalence alongside obesity and metabolic syndrome. Despite the promise of existing therapies such as thyroid hormone receptor-β (THR-β) agonists, PPAR agonists, FXR agonists, and GLP-1 receptor agonists, their effectiveness is limited by the complexity of the metabolic, inflammatory, and fibrotic pathways that drive MASLD progression, encompassing steatosis, metabolic dysfunction-associated steatohepatitis (MASH), and reversible liver fibrosis. Recent advances in targeted therapeutics, including RNA interference (RNAi), mRNA-based gene therapies, monoclonal antibodies, proteolysis-targeting chimeras (PROTAC), peptide-based strategies, cell-based therapies such as CAR-modified immune cells and stem cells, and extracellular vesicle-based approaches, have emerged as promising interventions. Alongside these developments, innovative drug delivery systems are being actively researched to enhance the stability, precision, and therapeutic efficacy of these biotherapeutics. These delivery strategies aim to optimize biodistribution, improve target-specific action, and reduce systemic exposure, thus addressing critical limitations of existing treatment modalities. This review provides a comprehensive exploration of the underlying biological mechanisms of MASLD and evaluates the potential of these cutting-edge biotherapeutics in synergy with advanced delivery approaches to address unmet clinical needs. By integrating fundamental disease biology with translational advancements, it aims to highlight future directions for the development of effective, targeted treatments for MASLD and its associated complications.
Collapse
Affiliation(s)
- Juhyeong Hong
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research Hanyang University, 04763 Seoul, South Korea; Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 04763 Seoul, South Korea
| | - Yong-Hee Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research Hanyang University, 04763 Seoul, South Korea; Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 04763 Seoul, South Korea; Cursus Bio Inc., Icure Tower, Gangnam-gu, Seoul 06170, Republic of Korea.
| |
Collapse
|
6
|
Zhang Y, Li J, Wang Z, Chen J, Zhao M, Guo C, Wang T, Li R, Zhang H, Ma X, Wen Y, Zeng J, Efferth T. Preclinical evidence construction for epigallocatechin-3-gallate against non-alcoholic fatty liver disease: a meta-analysis and machine learning study. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156651. [PMID: 40327947 DOI: 10.1016/j.phymed.2025.156651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/20/2024] [Accepted: 03/14/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) has emerged as a significant health concern worldwide, exhibiting an increasing incidence that necessitates immediate intervention. Epigallocatechin-3-gallate (EGCG) has shown significant pharmacological benefits for liver diseases, including NAFLD. However, its efficacy in this context has not been systematically evaluated. PURPOSE This meta-analysis aimed to consolidate preclinical evidence on the effectiveness and mechanisms of EGCG in treating NAFLD. METHODS We conducted a comprehensive literature search for preclinical studies from the inception of each database to April 2024, including Excerpta Medica Database, PubMed, Web of Science, Cochrane Library, China National Knowledge Infrastructure, Wanfang, and China Science and Technology Journal Database. These studies were manually screened based on predefined criteria. Data extraction was followed by pooled effect size calculations using Stata 16.0. A machine learning approach was also utilized to examine the temporal relationships among variables. RESULTS Seventeen studies, involving 293 animals, were analyzed. Our analysis indicates that EGCG significantly reduces ALT, AST, hepatic triglyceride, serum TG, hepatic TC, serum TC. The targets of EGCG may include antioxidants, regulation of lipid metabolism, anti-inflammation, improvement of insulin resistance, and inhibition of hepatic fibrosis. EGCG exerted its effects on NAFLD by modulating key signaling pathways, including PI3K/Akt/AMPK, TGF-β/Smad, Nrf2, NF-κB, and ROS/MAPK, highlighting its multifaceted mechanisms of action. The machine learning methods employed to ascertain the temporal relationship between the intervention and the outcome indicated that the optimal duration of the intervention was 10 to 15 weeks. CONCLUSIONS The efficacy of EGCG in treating NAFLD has been predicted within a time frame of 10-15 weeks. It may exert its effects primarily through the NF-κB and Nrf2 pathways, which regulate the ROS phenotype. EGCG may represent a promising target for the treatment of NAFLD.
Collapse
Affiliation(s)
- Yuanhao Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jianguo Li
- Bazhong Hospital of Traditional Chinese Medicine, Bazhong, China
| | - Zexin Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Maoyuan Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Cui Guo
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Tingyao Wang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Ruilin Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Hebin Zhang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Xiao Ma
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yueqiang Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jinhao Zeng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
7
|
Huang YD, Zhao XL, Lin Y, Ouyang XM, Cheng XS, Liang LY, Huo YN, Xie GJ, Lin JH, Jazag A, Guleng B. Mindin orchestrates the macrophage-mediated resolution of liver fibrosis in mice. Hepatol Int 2025:10.1007/s12072-025-10813-7. [PMID: 40186763 DOI: 10.1007/s12072-025-10813-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/28/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND & AIMS Liver disease that progresses to cirrhosis is an enormous health problem worldwide. The extracellular matrix protein Mindin is known to have immune functions, but its role in liver homeostasis remains largely unexplored. We aimed to characterize the role of Mindin in the regulation of liver fibrosis. APPROACH & RESULTS Mindin was upregulated in mice with carbon tetrachloride (CCl4) or thioacetamide (TAA)-induced liver fibrosis, and was primarily expressed in hepatocytes. Global Mindin knockout mice were generated, which were susceptible to liver fibrosis. Notably, Mindin failed to activate hepatic stellate cells directly; however, it played a role in promoting the recruitment and phagocytosis of macrophages, and caused a phenotypic switch toward restorative macrophages during liver fibrosis. Furthermore, Mindin was found to bind to the αM-I domain of CD11b/CD18 heterodimeric receptors. To further explore this mechanism, we created Mindin and CD11b double-knockout (DKO) mice. In DKO mice, phagocytosis was further reduced, and liver fibrosis was markedly exacerbated. CONCLUSIONS Mindin promotes the resolution of liver fibrosis and the Mindin/CD11b axis might represent a novel target for the macrophage-mediated regression of liver fibrosis.
Collapse
Affiliation(s)
- Yong-Dong Huang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361004, China
| | - Xian-Ling Zhao
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361004, China
| | - Ying Lin
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361004, China
| | - Xiao-Mei Ouyang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361004, China
| | - Xiao-Shen Cheng
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361004, China
| | - Lai-Ying Liang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361004, China
| | - Ya-Ni Huo
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361004, China
| | - Gui-Jing Xie
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361004, China
| | - Jun-Hui Lin
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361004, China
| | - Amarsanaa Jazag
- Department of Medicine, Otoch Manramba University, Ulaanbaatar, Mongolia
| | - Bayasi Guleng
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361004, China.
- Department of Digestive Disease & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen, 361004, China.
| |
Collapse
|
8
|
Wang Y, Sanyal AJ, Hylemon P, Ren S. Discovery of a novel regulator, 3β-sulfate-5-cholestenoic acid, of lipid metabolism and inflammation. Am J Physiol Endocrinol Metab 2025; 328:E543-E554. [PMID: 40047198 DOI: 10.1152/ajpendo.00426.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/04/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025]
Abstract
Mitochondrial oxysterols, cholestenoic acid (CA), 25-hydroxycholesterol (25HC), and 27-hydroxycholesterol (27HC), are potent regulators involved in many important biological events. This study aimed to investigate the metabolic pathways of these oxysterols and their roles between hepatocytes and macrophages. LC-MS/MS analysis showed a novel regulatory molecule, 3β-sulfate-5-cholestenoic acid (3SCA), following the addition of CA in media culturing hepatocytes. Further study showed that 3SCA could also be derived from 27HC. In comparison, 25HC was converted to 25HC3S, which mostly remained in the cells and nuclei. The functional study showed that 3SCA significantly downregulated the expression of genes involved in lipid metabolism in hepatocytes and suppressed gene expression of proinflammatory cytokines induced by lipopolysaccharide in human macrophages. Based on the results, we conclude that 3SCA acts as a secretory regulator for the regulation of lipid metabolism and inflammatory responses in hepatocytes and macrophages. These findings shed light on understanding the unique metabolic pathways of these oxysterols and their possible roles in liver tissues.NEW & NOTEWORTHY This study identifies a novel oxysterol metabolite, 3β-sulfate-5-cholestenoic acid (3SCA), secreted by hepatocytes, which regulates lipid metabolism and inflammatory responses in hepatocytes and macrophages. These findings reveal previously unknown metabolic pathways of mitochondrial oxysterols and their roles in the progression and recovery of metabolic dysfunction-associated steatotic liver disease (MASLD), offering novel insights into potential therapeutic targets.
Collapse
Affiliation(s)
- Yaping Wang
- Department of Internal Medicine, Virginia Commonwealth University/McGuire VA Medical Center, Richmond, Virginia, United States
| | - Arun J Sanyal
- Department of Internal Medicine, Virginia Commonwealth University/McGuire VA Medical Center, Richmond, Virginia, United States
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Richmond, Virginia, United States
| | - Phillip Hylemon
- Department of Internal Medicine, Virginia Commonwealth University/McGuire VA Medical Center, Richmond, Virginia, United States
- Department of Microbiology, Virginia Commonwealth University/McGuire VA Medical Center, Richmond, Virginia, United States
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Richmond, Virginia, United States
| | - Shunlin Ren
- Department of Internal Medicine, Virginia Commonwealth University/McGuire VA Medical Center, Richmond, Virginia, United States
| |
Collapse
|
9
|
Castanho Martins M, Dixon ED, Lupo G, Claudel T, Trauner M, Rombouts K. Role of PNPLA3 in Hepatic Stellate Cells and Hepatic Cellular Crosstalk. Liver Int 2025; 45:e16117. [PMID: 39394864 PMCID: PMC11891384 DOI: 10.1111/liv.16117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 10/14/2024]
Abstract
AIMS Since its discovery, the patatin-like phospholipase domain containing 3 (PNPLA3) (rs738409 C>G p.I148M) variant has been studied extensively to unravel its molecular function. Although several studies proved a causal relationship between the PNPLA3 I148M variant and MASLD development and particularly fibrosis, the pathological mechanisms promoting this phenotype have not yet been fully clarified. METHODS We summarise the latest data regarding the PNPLA3 I148M variant in hepatic stellate cells (HSCs) activation and macrophage biology or the path to inflammation-induced fibrosis. RESULTS Elegant but contradictory studies have ascribed PNPLA3 a hydrolase or an acyltransferase function. The PNPLA3 I148M results in hepatic lipid accumulation, which predisposes the hepatocyte to lipotoxicity and lipo-apoptosis, producing DAMPs, cytokines and chemokines leading to recruitment and activation of macrophages and HSCs, propagating fibrosis. Recent studies showed that the PNPLA3 I148M variant alters HSCs biology via attenuation of PPARγ, AP-1, LXRα and TGFβ activity and signalling. CONCLUSIONS The advent of refined techniques in isolating HSCs has made PNPLA3's direct role in HSCs for liver fibrosis development more apparent. However, many other mechanisms still need detailed investigations.
Collapse
Affiliation(s)
- Maria Castanho Martins
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive HealthUniversity College London, Royal Free CampusLondonUK
| | - Emmanuel Dauda Dixon
- Hans Popper Laboratory of Molecular Hepatology, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - Giulia Lupo
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive HealthUniversity College London, Royal Free CampusLondonUK
| | - Thierry Claudel
- Hans Popper Laboratory of Molecular Hepatology, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - Krista Rombouts
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive HealthUniversity College London, Royal Free CampusLondonUK
| |
Collapse
|
10
|
Yang L, Li P, Zhao J, Bai Z, Zeng G, Liu X, Zou B, Li J. CAT and CXCL8 are crucial cofactors for the progression of nonalcoholic steatohepatitis to hepatocellular carcinoma, the immune infiltration and prognosis of hepatocellular carcinoma. Discov Oncol 2025; 16:272. [PMID: 40053253 PMCID: PMC11889291 DOI: 10.1007/s12672-025-02051-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 03/04/2025] [Indexed: 03/10/2025] Open
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is a malignant tumour characterized by high morbidity and mortality. Immunotherapy is an important treatment newly approved for the treatment for advanced hepatocellular carcinoma. However, how NASH progresses to HCC and the association between the immune signature in HCC and patient prognosis remain unclear. METHODS Data from NASH and NASH-HCC patients were obtained from the GEO database. Differentially expressed genes were screened and hub genes were identified. The enrichment analysis, clustering, cibersort, ssGSEA, Xcell and immune checkpoint expression data of the samples were analysed. Survival analysis of dual genes was performed using TCGA liver cancer samples and the lasso regression model, and Cox regression analysis was conducted. Pathology specimens from 21 NASH-associated hepatocellular carcinoma patients were collected, and immunohistochemical staining was used to verify gene expression. RESULTS Compared with HCC patients with high CAT and low CXCL8 expression, those with low CAT and high CXCL8 expression had significantly higher levels of infiltration of multiple immune cell types and the common immune checkpoints CD274, PDCD1 and CTLA4. Furthermore, CAT was a protective factor, and CXCL8 was a risk factor for the prognosis of HCC patients. CONCLUSION CAT and CXCL8 might impact NASH-HCC progression. HCC patients with low CAT and high CXCL8 expression might have more extensive immune cell infiltration and stronger tumour immune escape. However, probably due to their different effects on CD8 + T cells and reactive oxygen species, increased expression of CAT contributes to improved prognosis in HCC patients, whereas increased expression of CXCL8 leads to a poor prognosis.
Collapse
Affiliation(s)
- Liang Yang
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, 52 Mei Hua East Road, Zhuhai, 519000, Guangdong Province, China
| | - Peiping Li
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, 52 Mei Hua East Road, Zhuhai, 519000, Guangdong Province, China
| | - JiaLi Zhao
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, 52 Mei Hua East Road, Zhuhai, 519000, Guangdong Province, China
| | - Zirui Bai
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, 52 Mei Hua East Road, Zhuhai, 519000, Guangdong Province, China
| | - Guifang Zeng
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, 52 Mei Hua East Road, Zhuhai, 519000, Guangdong Province, China
| | - Xialei Liu
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, 52 Mei Hua East Road, Zhuhai, 519000, Guangdong Province, China.
| | - Baojia Zou
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, 52 Mei Hua East Road, Zhuhai, 519000, Guangdong Province, China.
| | - Jian Li
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, 52 Mei Hua East Road, Zhuhai, 519000, Guangdong Province, China.
| |
Collapse
|
11
|
Liu Z, Zhang X, Ben T, Li M, Jin Y, Wang T, Song Y. Focal adhesion in the tumour metastasis: from molecular mechanisms to therapeutic targets. Biomark Res 2025; 13:38. [PMID: 40045379 PMCID: PMC11884212 DOI: 10.1186/s40364-025-00745-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/11/2025] [Indexed: 03/09/2025] Open
Abstract
The tumour microenvironment is the "hotbed" of tumour cells, providing abundant extracellular support for growth and metastasis. However, the tumour microenvironment is not static and is constantly remodelled by a variety of cellular components, including tumour cells, through mechanical, biological and chemical means to promote metastasis. Focal adhesion plays an important role in cell-extracellular matrix adhesion. An in-depth exploration of the role of focal adhesion in tumour metastasis, especially their contribution at the biomechanical level, is an important direction of current research. In this review, we first summarize the assembly of focal adhesions and explore their kinetics in tumour cells. Then, we describe in detail the role of focal adhesion in various stages of tumour metastasis, especially its key functions in cell migration, invasion, and matrix remodelling. Finally, we describe the anti-tumour strategies targeting focal adhesion and the current progress in the development of some inhibitors against focal adhesion proteins. In this paper, we summarize for the first time that focal adhesion play a positive feedback role in pro-tumour metastatic matrix remodelling by summarizing the five processes of focal adhesion assembly in a multidimensional way. It is beneficial for researchers to have a deeper understanding of the role of focal adhesion in the biological behaviour of tumour metastasis and the potential of focal adhesion as a therapeutic target, providing new ideas for the prevention and treatment of metastases.
Collapse
Affiliation(s)
- Zonghao Liu
- Department of Radiotherapy, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China
- The First Clinical College, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Xiaofang Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Tianru Ben
- The First Clinical College, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Mo Li
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China
| | - Yi Jin
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China
| | - Tianlu Wang
- Department of Radiotherapy, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China.
- Department of Radiotherapy, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning Province, 110042, People's Republic of China.
- Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning Province, 116024, P. R. China.
| | - Yingqiu Song
- Department of Radiotherapy, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China.
- Department of Radiotherapy, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China.
| |
Collapse
|
12
|
Hawley J, Tang Y, Sjöström A, Fuentes-Alburo A, Tranquart F. The Clinical Utility of Liver-Specific Ultrasound Contrast Agents During Hepatocellular Carcinoma Imaging. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:415-427. [PMID: 39674715 DOI: 10.1016/j.ultrasmedbio.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 12/16/2024]
Abstract
Hepatocellular carcinoma (HCC) is the most common form of hepatic malignancy, with high mortality rates recorded globally. Early detection through clinical biomarkers, medical imaging and histological assessment followed by rapid intervention are integral for positive patient outcomes. Although contrast-enhanced computed tomography scans and magnetic resonance imaging are recognised as the reference standard for the diagnosis and staging of HCC in international guidelines, ultrasound (US) examination is recommended as a screening tool for patients at risk. Contrast-enhanced US (CEUS) elevates the standard of an US examination using US contrast agents (UCAs), capable of diagnosing focal liver lesions with high efficacy. Most UCAs are purely intravascular, offering clinicians a dynamic representation of a lesions' arterial phase vascular kinetics, which is seldom seen in such detail during computed tomography or magnetic resonance imaging assessments. Despite its benefits, there is incongruity between international societies on the role of CEUS in the HCC clinical pathway. The transient nature of pure blood-pool agents is suggested to be insufficient to justify CEUS as a primary modality due to the inability to consistently perform whole liver imaging, alongside disputes regarding its capabilities to differentiate HCC from intrahepatic cholangiocarcinoma. A sinusoidal phase UCA affords clinicians the opportunity to perform whole liver imaging through Kupffer cell uptake in addition to visualising lesion vascular kinetics in the arterial and portal venous phases. Therefore, the purpose of this review was to examine the role of CEUS in the HCC clinical pathway in its current practice and observe how a Kupffer cell sinusoidal phase UCA may supplement contemporary diagnostic techniques through a multi-modality, multi-agent approach.
Collapse
Affiliation(s)
- Joshua Hawley
- GE HealthCare Pharmaceutical Diagnostics, Chalfont St. Giles, UK; Chesterfield Royal Hospital Foundation NHS Trust, Chesterfield, UK.
| | - Yongqing Tang
- GE HealthCare Pharmaceutical Diagnostics, Chalfont St. Giles, UK
| | - Anders Sjöström
- GE HealthCare Pharmaceutical Diagnostics, Chalfont St. Giles, UK
| | | | | |
Collapse
|
13
|
Mincheva G, Moreno-Manzano V, Felipo V, Llansola M. Extracellular vesicles from mesenchymal stem cells improve liver injury in rats with mild liver damage. Underlying mechanisms and role of TGFβ. Life Sci 2025; 364:123429. [PMID: 39884339 DOI: 10.1016/j.lfs.2025.123429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Preventing the progression of liver damage to fibrosis would be beneficial for patients with steatotic liver disease (SLD). Mesenchymal stem cells (MSC) are a promising therapy for SLD and derived extracellular vesicles (EVs) could even improve the treatment's efficacy and safety. However, the mechanisms of MSC-EVs beneficial effects are not well known. It has been suggested that modifying the EVs cargo could improve their beneficial effects. The aims of this study were to assess if MSC-EVs reduce liver damage in a rat model of mild liver damage; to analyze the underlying mechanisms and to assess if silencing TGFβ enhances the beneficial effects of MSC-EVs. CCl4 was injected three times per week during four weeks to induce mild liver damage. EVs from human adipocyte MSC and from TGFβ-depleted MSC (siTGFβ-MSC-EVs) were injected in the tail vein. Steatosis, fibrosis, liver inflammation, macrophage infiltration and liver content of fibrotic markers, DAMPs, cytokines and bile acids were analyzed. Normal MSC-EVs reduce the CCL2 increase in liver, macrophage infiltration and the increases in the fibrosis markers collagen I and α-SMA. Treatment with siTGFβ-MSC-EVs, in addition, reduces liver steatosis, the increase of bile acids (mainly TCA), and DAMP HMGB1 levels, inducing a larger reduction of collagen I in liver of CCl4 rats. Treatment with MSCs-EVs effectively reduces early liver damage. Silencing of TGFβ in MSCs enhances the beneficial effects by additional mechanisms. Early treatment with MSC-EVs, especially after silencing TGFβ, could improve liver damage in SLD patients.
Collapse
Affiliation(s)
- Gergana Mincheva
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Victoria Moreno-Manzano
- Laboratory of Neuronal and Tissue Regeneration, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Marta Llansola
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| |
Collapse
|
14
|
Chi KY, Kim G, Kim H, Kim H, Jo S, Lee J, Lee Y, Yoon H, Cho S, Kim J, Lee JS, Yeon GB, Kim DS, Park HJ, Kim JH. Optimization of culture conditions to generate vascularized multi-lineage liver organoids with structural complexity and functionality. Biomaterials 2025; 314:122898. [PMID: 39447308 DOI: 10.1016/j.biomaterials.2024.122898] [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: 06/03/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
Hepatic organoids (HOs), primarily composed of hepatobiliary cells, do not represent the pathogenesis of liver diseases due to the lack of non-parenchymal cells. Multi-lineage liver organoids (mLOs) containing various cell types found in the liver offer a promising in vitro disease model. However, their structural complexity remains challenging to achieve due to the difficulty in optimizing culture conditions that meet the growth need of all component cell types. Here, we demonstrate that cystic HOs generated from hPSCs can be expanded long-term and serve as a continuous source for generating complex mLOs. Assembling cystic HOs with hPSC-derived endothelial and hepatic stellate cell-like cells under conventional HO culture conditions failed to support the development of multiple cell types within mLOs, resulting in biased differentiation towards specific cell types. In contrast, modulating the cAMP/Wnt/Hippo signaling pathways with small molecules during assembly and differentiation phases efficiently generate mLOs containing both hepatic parenchymal and non-parenchymal cells. These mLOs exhibited structural complexity and functional maturity, including vascular network formation between parenchymal lobular structures, cell polarity for bile secretion, and the capacity to respond to fibrotic stimuli. Our study underscores the importance of modulating signaling pathways to enhance mLO structural complexity for applications in modeling liver pathologies.
Collapse
Affiliation(s)
- Kyun Yoo Chi
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Gyeongmin Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Hyojin Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Hyemin Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, South Korea
| | - Seongyea Jo
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, South Korea
| | - Jihun Lee
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Youngseok Lee
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea; Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Heeseok Yoon
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Seunghyun Cho
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Jeongjun Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Jin-Seok Lee
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Gyu-Bum Yeon
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, South Korea; Laboratory of Reprogramming and Differentiation, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Dae-Sung Kim
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, South Korea; Laboratory of Reprogramming and Differentiation, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Han-Jin Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, South Korea
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea.
| |
Collapse
|
15
|
Zimmermann A, Scheffschick A, Hänsel R, Borchardt H, Liu JL, Ehnert S, Schicht G, Seidemann L, Aigner A, Schiffmann S, Nüssler A, Seehofer D, Damm G. A new human autologous hepatocyte/macrophage co-culture system that mimics drug-induced liver injury-like inflammation. Arch Toxicol 2025; 99:1167-1185. [PMID: 39710784 PMCID: PMC11821741 DOI: 10.1007/s00204-024-03943-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/11/2024] [Indexed: 12/24/2024]
Abstract
The development of in vitro hepatocyte cell culture systems is crucial for investigating drug-induced liver injury (DILI). One prerequisite for monitoring DILI related immunologic reactions is the extension of primary human hepatocyte (PHH) cultures towards the inclusion of macrophages. Therefore, we developed and characterized an autologous co-culture system of PHH and primary human hepatic macrophages (hepM) (CoC1). We compared CoC1 with a co-culture of the same PHH batch + M0 macrophages derived from THP1 cells (CoC2) in order to represent a donor independent macrophage reaction. Then, we treated the mono- and co-cultures with drugs that cause DILI-menadione (MEN, 1 or 10 µM, 3 h), diclofenac (DIC, 0.5 or 5 mM, 6 h), or acetaminophen (APAP, 0.5 or 5 mM, 6 h)-and assessed culture stability, cell activity, macrophage differentiation, cytokine production and cell viability. Without drug treatment, CoC1 was the most stable over a culture time of up to 60 h. Cytokine array analysis revealed a proinflammatory profile of PHH mono-cultures due to isolation stress but showed different influences of hepM and M0 on the cytokine profile in the co-cultures. MEN, DIC and APAP treatment led to donor-dependent signs of cell stress and toxicity. HepM can either promote or reduce the DILI effects donor dependently in CoC1. CoC2 are slightly less sensitive than CoC1 in representing DILI. In summary, we present a new autologous co-culture system that can mimic DILI in a donor-dependent manner. This cellular system could be useful for new drug testing strategies and reducing animal testing.
Collapse
Affiliation(s)
- Andrea Zimmermann
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic and Polyclinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Germany
| | - Andrea Scheffschick
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic and Polyclinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Germany
| | - René Hänsel
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic and Polyclinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, Leipzig, Germany
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), Leipzig University, Leipzig, Germany
| | - Hannes Borchardt
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Jia Li Liu
- Department of General, Visceral- and Transplantation Surgery, Charité - University Medicine Berlin, Berlin, Germany
| | - Sabrina Ehnert
- Department of Traumatology, BG Trauma Center, University of Tübingen, Tübingen, Germany
| | - Gerda Schicht
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic and Polyclinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Germany
| | - Lena Seidemann
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic and Polyclinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, Leipzig, Germany
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Susanne Schiffmann
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt Am Main, Germany
| | - Andreas Nüssler
- Department of Traumatology, BG Trauma Center, University of Tübingen, Tübingen, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic and Polyclinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Germany
- Department of General, Visceral- and Transplantation Surgery, Charité - University Medicine Berlin, Berlin, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic and Polyclinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, Leipzig, Germany.
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Germany.
- Department of General, Visceral- and Transplantation Surgery, Charité - University Medicine Berlin, Berlin, Germany.
| |
Collapse
|
16
|
Sarawi WS, Attia HA, Alzoubi A, Alanazi N, Mohammad R, Ali RA. Cardamom extract alleviates tamoxifen-induced liver damage by suppressing inflammation and pyroptosis pathway. Sci Rep 2025; 15:4744. [PMID: 39922887 PMCID: PMC11807216 DOI: 10.1038/s41598-025-89091-0] [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: 08/22/2024] [Accepted: 02/03/2025] [Indexed: 02/10/2025] Open
Abstract
Tamoxifen (TAM) is extensively used to manage estrogen receptor-positive breast cancer. Despite its effectiveness, its administration can negatively impact various organs, including the liver. This research focused on the effects of TAM on the pyroptotic pathway in the liver and evaluated the potential of cardamom extract (CRDE) to lessen hepatic damage of TAM in female rats. Rats received 45 mg/kg of TAM injections for 10 days, while the groups treated with CRDE received 12 ml/kg of CRDE for 20 days, commencing 10 days before TAM administration. TAM exposure resulted in apparent degenerations in hepatic tissue with inflammatory cell infiltration and loss of architectures. Serum levels of liver enzymes including alanine aminotransferase, aspartate aminotransferase and alkaline phosphatase were elevated, along with hepatic oxidative stress, as shown by increased lipid peroxidation with lower levels of reduced glutathione. TAM caused inflammation in the liver tissue as indicated by higher levels of tumor necrosis factor-α and interleukin-6 as well as increased expression of CD68; a phagocytic Kupffer's cells marker. Additionally, the protein expression analysis revealed a high expression of pyroptotic markers including NLRP3-inflammasome, caspase-1, and gasdermin D. Conversely, CRDE treatment effectively neutralized the biochemical, histological, and protein expression alterations induced by TAM. In conclusion, CRDE demonstrated the potential to protect the liver from TAM-induced damage by regulating mechanisms involving oxidative damage, inflammation, and pyroptosis.
Collapse
Affiliation(s)
- Wedad S Sarawi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia.
| | - Hala A Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia
| | - Afraa Alzoubi
- Department of Bioengineering, Imperial College London, London, UK
| | - Nour Alanazi
- College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia
| | - Raeesa Mohammad
- Department of Histology, College of Medicine, King Saud University, P.O. Box 2925, Riyadh, 11461, Saudi Arabia
| | - Rehab A Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia
| |
Collapse
|
17
|
Young DJ, Edwards AJ, Quiroz Caceda KG, Liberzon E, Barrientos J, Hong SG, Turner J, Choyke PL, Arlauckas S, Lazorchak AS, Morgan RA, Sato N, Dunbar CE. In vivo tracking of ex-vivo-generated 89Zr-oxine-labeled plasma cells by PET in a non-human primate model. Mol Ther 2025; 33:580-594. [PMID: 39741408 PMCID: PMC11852699 DOI: 10.1016/j.ymthe.2024.12.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 11/12/2024] [Accepted: 12/27/2024] [Indexed: 01/03/2025] Open
Abstract
B cells are an attractive platform for engineering to produce protein-based biologics absent in genetic disorders, and potentially for the treatment of metabolic diseases and cancer. As part of pre-clinical development of B cell medicines, we demonstrate a method to collect, ex vivo expand, differentiate, radioactively label, and track adoptively transferred non-human primate (NHP) B cells. These cells underwent 10- to 15-fold expansion, initiated IgG class switching, and differentiated into antibody-secreting cells. Zirconium-89-oxine-labeled cells were infused into autologous donors without any preconditioning and tracked by PET/CT imaging. Within 24 h of infusion, 20% of the initial dose homed to the bone marrow and spleen and distributed stably and equally between the two. Interestingly, approximately half of the dose homed to the liver. Image analysis of the bone marrow demonstrated inhomogeneous distribution of the cells. The subjects experienced no clinically significant side effects or laboratory abnormalities. A second infusion of B cells into one of the subjects resulted in an almost identical distribution of cells, suggesting possibly a non-limiting engraftment niche and feasibility of repeated infusions. This work supports the NHP as a valuable model to assess the potential of B cell medicines as potential treatment for human diseases.
Collapse
Affiliation(s)
- David J Young
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | - Kevin G Quiroz Caceda
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | - So Gun Hong
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Peter L Choyke
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | - Noriko Sato
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cynthia E Dunbar
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Mo H, Yue P, Li Q, Tan Y, Yan X, Liu X, Xu Y, Luo Y, Palihati S, Yi C, Zhang H, Yuan M, Yang B. The role of liver sinusoidal endothelial cells in metabolic dysfunction-associated steatotic liver diseases and liver cancer: mechanisms and potential therapies. Angiogenesis 2025; 28:14. [PMID: 39899173 DOI: 10.1007/s10456-025-09969-5] [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: 11/25/2024] [Accepted: 01/15/2025] [Indexed: 02/04/2025]
Abstract
Liver sinusoidal endothelial cells (LSECs), with their unique morphology and function, have garnered increasing attention in chronic liver disease research. This review summarizes the critical roles of LSECs under physiological conditions and in two representative chronic liver diseases: metabolic dysfunction-associated steatotic liver disease (MASLD) and liver cancer. Under physiological conditions, LSECs act as selective barriers, regulating substance exchange and hepatic blood flow. Interestingly, LSECs exhibit contrasting roles at different stages of disease progression: in the early stages, they actively resist disease advancement and help restore sinusoidal homeostasis; whereas in later stages, they contribute to disease worsening. During this transition, LSECs undergo capillarization, lose their characteristic markers, and become dysfunctional. As the disease progresses, LSECs closely interact with hepatocytes, hepatic stellate cells, various immune cells, and tumor cells, driving processes such as steatosis, inflammation, fibrosis, angiogenesis, and carcinogenesis. Consequently, targeting LSECs represents a promising therapeutic strategy for chronic liver diseases. Relevant therapeutic targets and potential drugs are summarized in this review.
Collapse
Affiliation(s)
- Hanjun Mo
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Pengfei Yue
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Qiaoqi Li
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Yinxi Tan
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
| | - Xinran Yan
- Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyue Liu
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuanwei Xu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yingzhe Luo
- Department of Medical Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Chengdu, 610075, Sichuan, China
| | - Suruiya Palihati
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Cheng Yi
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| | - Hua Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, No. 20, Section 3, South Renmin Road, Chengdu, 610041, China.
- Key Laboratory of Chronobiology (Sichuan University), National Health Commission of China, Chengdu, 610041, China.
| | - Minlan Yuan
- Mental Health Center and Psychiatric Laboratory, The State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China.
| | - Biao Yang
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
19
|
Hu Y, Schnabl B, Stärkel P. Origin, Function, and Implications of Intestinal and Hepatic Macrophages in the Pathogenesis of Alcohol-Associated Liver Disease. Cells 2025; 14:207. [PMID: 39936998 PMCID: PMC11816606 DOI: 10.3390/cells14030207] [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: 11/15/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 02/13/2025] Open
Abstract
Macrophages are members of the human innate immune system, and the majority reside in the liver. In recent years, they have been recognized as essential players in the maintenance of liver and intestinal homeostasis as well as key guardians of their respective immune systems, and they are increasingly being recognized as such. Paradoxically, they are also likely involved in chronic pathologies of the gastrointestinal tract and potentially in the alteration of the gut-liver axis in alcohol use disorder (AUD) and alcohol-associated liver disease (ALD). To date, the causal relationship between macrophages, the pathogenesis of ALD, and the immune dysregulation of the gut remains unclear. In this review, we will discuss our current understanding of the heterogeneity of intestinal and hepatic macrophages, their ontogeny, the potential factors that regulate their origin, and the evidence of how they are associated with the manifestation of chronic inflammation. We will also illustrate how the micro-environment of the intestine shapes the phenotypes and functionality of the macrophage compartment in both the intestines and liver and how they change during chronic alcohol abuse. Finally, we highlight the obstacles to current research and the prospects for this field.
Collapse
Affiliation(s)
- Yifan Hu
- Laboratory of Hepato-Gastroenterology, Institute of Clinical and Experimental Research, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA 92161, USA;
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Peter Stärkel
- Laboratory of Hepato-Gastroenterology, Institute of Clinical and Experimental Research, Université Catholique de Louvain, 1200 Brussels, Belgium;
- Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
20
|
Qian Y, Zhao J, Wu H, Kong X. Innate immune regulation in inflammation resolution and liver regeneration in drug-induced liver injury. Arch Toxicol 2025; 99:115-126. [PMID: 39395921 DOI: 10.1007/s00204-024-03886-0] [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] [Accepted: 10/02/2024] [Indexed: 10/14/2024]
Abstract
Drug-induced liver injury (DILI) is an acute liver injury that poses a significant threat to human health. In severe cases, it can progress into chronic DILI or even lead to liver failure. DILI is typically caused by either intrinsic hepatotoxicity or idiosyncratic metabolic or immune responses. In addition to the direct damage drugs inflict on hepatocytes, the immune responses and liver inflammation triggered by hepatocyte death can further exacerbate DILI. Initially, we briefly discussed the differences in immune cell activation based on the type of liver cell death (hepatocytes, cholangiocytes, and LSECs). We then focused on the role of various immune cells (including macrophages, monocytes, neutrophils, dendritic cells, liver sinusoidal endothelial cells, eosinophils, natural killer cells, and natural killer T cells) in both the liver injury and liver regeneration stages of DILI. This article primarily reviews the role of innate immune regulation mediated by these immune cells in resolving inflammation and promoting liver regeneration during DILI, as well as therapeutic approaches targeting these immune cells for the treatment of DILI. Finally, we discussed the activation and function of liver progenitor cells (LPCs) during APAP-induced massive hepatic necrosis and the involvement of chronic inflammation in DILI.
Collapse
Affiliation(s)
- Yihan Qian
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai, China
| | - Jie Zhao
- Department of Liver Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hailong Wu
- Shanghai Key Laboratory of Molecular Imaging, Collaborative Innovation Center for Biomedicines, Shanghai University of Medicine and Health Sciences, Shanghai, China.
| | - Xiaoni Kong
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai, China.
| |
Collapse
|
21
|
Zhang J, Li N, Hu X. Metabolic Reprograming of Macrophages: A New Direction in Traditional Chinese Medicine for Treating Liver Failure. J Immunol Res 2024; 2024:5891381. [PMID: 39741958 PMCID: PMC11688140 DOI: 10.1155/jimr/5891381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 09/03/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
Acute liver failure (ALF) is a fulminant clinical syndrome that usually leads to multiple organ failure and high mortality. Macrophages play a crucial role in the initiation, development, and recovery of ALF. Targeting macrophages through immunotherapy holds significant promise as a therapeutic strategy. These cells exhibit remarkable plasticity, enabling them to differentiate into various subtypes based on changes in their surrounding microenvironment. M1-type macrophages are associated with a pro-inflammatory phenotype and primarily rely predominantly on glycolysis. In contrast, M2-type macrophages, which are characterized by anti-inflammatory phenotype, predominantly obtain their energy from oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO). Shifting macrophage metabolism from glycolysis to OXPHOS inhibits M1 macrophage activation and promotes M2 macrophage activation, thereby exerting anti-inflammatory and reparative effects. This study elucidates the relationship between macrophage activation and glucose metabolism reprograming from an immunometabolism perspective. A comprehensive literature review revealed that several signaling pathways may regulate macrophage polarization through energy metabolism, including phosphatidyl-inositol 3-kinase/protein kinase B (PI3K/AKT), mammalian target of rapamycin (mTOR)/hypoxia-inducible factor 1α (HIF-1α), nuclear factor-κB (NF-κB), and AMP-activated protein kinase (AMPK), which exhibit crosstalk with one another. Additionally, we systematically reviewed several traditional Chinese medicine (TCM) monomers that can modulate glucose metabolism reprograming and influence the polarization states of M1 and M2 macrophages. This review aimed to provide valuable insights that could contribute to the development of new therapies or drugs for ALF.
Collapse
Affiliation(s)
- Junli Zhang
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Na Li
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyu Hu
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
22
|
Rabas N, Ferreira RMM, Di Blasio S, Malanchi I. Cancer-induced systemic pre-conditioning of distant organs: building a niche for metastatic cells. Nat Rev Cancer 2024; 24:829-849. [PMID: 39390247 DOI: 10.1038/s41568-024-00752-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 10/12/2024]
Abstract
From their early genesis, tumour cells integrate with the surrounding normal cells to form an abnormal structure that is tightly integrated with the host organism via blood and lymphatic vessels and even neural associations. Using these connections, emerging cancers send a plethora of mediators that efficiently perturb the entire organism and induce changes in distant tissues. These perturbations serendipitously favour early metastatic establishment by promoting a more favourable tissue environment (niche) that supports the persistence of disseminated tumour cells within a foreign tissue. Because the establishment of early metastatic niches represents a key limiting step for metastasis, the creation of a more suitable pre-conditioned tissue strongly enhances metastatic success. In this Review, we provide an updated view of the mechanisms and mediators of primary tumours described so far that induce a pro-metastatic conditioning of distant organs, which favours early metastatic niche formation. We reflect on the nature of cancer-induced systemic conditioning, considering that non-cancer-dependent perturbations of tissue homeostasis are also able to trigger pro-metastatic conditioning. We argue that a more holistic view of the processes catalysing metastatic progression is needed to identify preventive or therapeutic opportunities.
Collapse
Affiliation(s)
- Nicolas Rabas
- Tumour-Host Interaction Laboratory, The Francis Crick Institute, London, UK
| | - Rute M M Ferreira
- Tumour-Host Interaction Laboratory, The Francis Crick Institute, London, UK
| | - Stefania Di Blasio
- Tumour-Host Interaction Laboratory, The Francis Crick Institute, London, UK
| | - Ilaria Malanchi
- Tumour-Host Interaction Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
23
|
Hsieh MH, Kao TY, Hsieh TH, Kao CC, Peng CY, Lai HC, Cheng HH, Ho MW, Chi CY, Kao JT. Predictors of liver fibrosis changes assessed by paired liver biopsies in chronic hepatitis C patients treated with direct-acting antivirals. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2024; 57:840-853. [PMID: 39216998 DOI: 10.1016/j.jmii.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/15/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND/PURPOSE There are limited studies performing paired liver biopsies in chronic hepatitis C (CHC) patients treated with direct-acting antivirals (DAA). We aimed to investigate the predictors of liver fibrosis changes assessed by paired liver biopsies in these patients. METHODS From March 2017 to March 2020, 113 CHC patients were prospectively enrolled to receive DAA therapy at our hospital. Paired liver biopsies were performed at baseline and 12 weeks after the end of treatment. RESULTS Among the entire cohort, the rate of sustained virological response (SVR) was 100%. Four baseline variables independently predicted fibrosis regression, including age <65 years [odds ratio (OR) = 2.725, p = 0.036], fibrosis stages (METAVIR scores) < 3 (OR = 4.874, p = 0.040), hemoglobin levels ≥12.5 g/dL (OR = 3.538, p = 0.029), and platelet counts ≥160 103/μL (OR = 2.958, p = 0.023). Besides, five independent predictors of fibrosis progression included baseline age ≥66 years (OR = 16.351, p = 0.024), body mass index (BMI) ≥26.5 kg/m2 (OR = 21.666, p = 0.009), sofosbuvir/ribavirin use (OR = 29.465, p = 0.031), platelet counts <119 103/μL (OR = 33.739, p = 0.026), and the absence of alanine aminotransferase (ALT) levels declining from >35 U/L at baseline to ≤35 U/L at 4 weeks after baseline (OR = 284.534, p = 0.026). CONCLUSION For DAA-treated CHC patients, those with baseline age <65 years, fibrosis stages <3, hemoglobin levels ≥12.5 g/dL, or platelet counts ≥160 103/μL are more likely to attain fibrosis regression. There is a higher risk of fibrosis progression in those with baseline age ≥66 years, BMI ≥26.5 kg/m2, sofosbuvir/ribavirin use, platelet counts <119 103/μL, or the absence of early ALT normalization at 4 weeks after baseline.
Collapse
Affiliation(s)
- Ming-Han Hsieh
- Department of Medicine, School of Medicine, China Medical University, Taichung, Taiwan; Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Tzu-Yu Kao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ting-Hui Hsieh
- Department of Pharmacy, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Chi Kao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Cheng-Yuan Peng
- Department of Medicine, School of Medicine, China Medical University, Taichung, Taiwan; Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Hsueh-Chou Lai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Hsing-Hung Cheng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Mao-Wang Ho
- Department of Medicine, School of Medicine, China Medical University, Taichung, Taiwan; Division of Infectious Diseases, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Yu Chi
- Department of Medicine, School of Medicine, China Medical University, Taichung, Taiwan; Division of Infectious Diseases, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Jung-Ta Kao
- Department of Medicine, School of Medicine, China Medical University, Taichung, Taiwan; Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
24
|
Zeng G, Krishnamurthy S, Staats Pires A, Guller A, Chaganti J, Tun N, Lockart I, Montagnese S, Brew B, Guillemin GJ, Danta M, Heng B. Activation of the kynurenine pathway identified in individuals with covert hepatic encephalopathy. Hepatol Commun 2024; 8:e0559. [PMID: 39774873 PMCID: PMC11567712 DOI: 10.1097/hc9.0000000000000559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 07/24/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND HE is a neuropsychiatric complication of liver disease characterized by systemic elevation in ammonia and proinflammatory cytokines. These neurotoxins cross the blood-brain barrier and cause neuroinflammation, which can activate the kynurenine pathway (KP). This results in dysregulated production of neuroactive KP metabolites, such as quinolinic acid, which is known to cause astrocyte and neuronal death. Our aim was to compare KP activity between patients with covert HE (CHE), patients without encephalopathic cirrhosis (NHE), and healthy controls (HCs). METHODS This was a single-center prospective cohort study conducted between 2018 and 2021 at St Vincent's Hospital, Sydney. Overall, 13 patients with CHE, 10 patients with NHE, and 12 with HC were recruited. Patients with cirrhosis were diagnosed with CHE if they scored ≤-4 on the Psychometric Hepatic Encephalopathy Score. KP metabolite levels were quantified on plasma samples via HPLC and gas chromatography/mass spectrometry. One-way Kruskal-Wallis test was used to compare the expression levels of KP enzymes. RESULTS KP was highly activated in patients with cirrhosis, demonstrated by higher levels of activity in the rate-limiting enzymes, indoleamine 2,3-dioxygenase, and tryptophan-2,3-dioxygenase in both CHE (65.04±20.72, p=0.003) and patients with NHE (64.85±22.10, p=0.015) compared to HC (40.95±7.301). Higher quinolinic acid concentrations were demonstrated in CHE (3726 nM±3385, p<0.001) and patients with NHE (1788 nM±632.3, p=0.032) compared to HC (624 nM±457). KP activation was positively correlated with inflammatory marker C-reactive protein in patients with CHE (Rs=0.721, p≤0.01). CONCLUSIONS KP is highly activated in patients with CHE, resulting in heightened production of neurotoxic metabolites. Dysregulation of the pathway is demonstrable in patients who do not yet show clinical signs of neurocognitive impairment. Therapeutic agents that modulate KP activity may be able to alleviate symptoms of patients with CHE.
Collapse
Affiliation(s)
- Georgia Zeng
- Department of Gastroenterology and Hepatology, St Vincent’s Hospital, Sydney, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Campus, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Shivani Krishnamurthy
- Macquarie Medicine School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Ananda Staats Pires
- Macquarie Medicine School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Anna Guller
- Macquarie Medicine School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Computational Neurosurgery (CNS) Laboratory, Macquarie Medicine School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Joga Chaganti
- Department of Medical Imaging, St Vincent’s Hospital, Sydney, Australia
| | - Nway Tun
- Department of Gastroenterology and Hepatology, St Vincent’s Hospital, Sydney, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Campus, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Ian Lockart
- Department of Gastroenterology and Hepatology, St Vincent’s Hospital, Sydney, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Campus, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Sara Montagnese
- Department of Medicine, Padova University Hospital, Padova, Italy
- Department of Chronobiology, Institute for Sustainability, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Bruce Brew
- Department of Neurology, St Vincent’s Hospital, Sydney, Australia
| | | | - Mark Danta
- Department of Gastroenterology and Hepatology, St Vincent’s Hospital, Sydney, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Campus, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Benjamin Heng
- Macquarie Medicine School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| |
Collapse
|
25
|
Ruathong W, Khuituan P, Peerakietkhajorn S, Teanpaisan R, Nopparat J. The probiotic Lacticaseibacillus rhamnosus SD11 alleviates the progression of liver and colon damage through modulation of inflammation and tight junction proteins in streptozotocin-induced diabetic mice. PLoS One 2024; 19:e0313395. [PMID: 39570868 PMCID: PMC11581286 DOI: 10.1371/journal.pone.0313395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024] Open
Abstract
Lacticaseibacillus rhamnosus SD11 (SD11) has several health benefits for the host, including antidiabetic, anti-inflammatory, and antimicrobial effects. However, the antidiabetic mechanism of SD11 has not been clearly elucidated. The current study assessed the effects of SD11 and the associated underlying mechanisms on streptozotocin (STZ)-induced diabetic mice. Compared with the normal control, SD11 supplementation for 4 weeks significantly improved the metabolic profiles, including body weight (BW), fasting blood glucose (FBG), fasting insulin level (FIN), and liver index (LI), in conjunction with a lower NAS score. A notable reduction in the liver function parameters aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP) and total cholesterol (TC), together with histopathology studies, supported diabetic recovery by SD11. A closer examination of two major markers for the insulin pathway, insulin receptor (INSR) and insulin substrate (IRS)-1, revealed that SD11 could exert its glucose control through the upregulation of these molecules, which were almost demolished in nontreated diabetic livers. Additionally, SD11-treated mice exhibited alleviation of oxidative stress enzymes; downregulation of proinflammatory cytokines, including interleukin (IL)-6, IL-1β, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ; and decreased infiltration of macrophages into liver tissue. These findings were concomitant with the preservation of the tight junction proteins occludin and zona occludin (ZO)-1, which in turn lowered the levels of the inflammatory cytokines IL-1β and TNF-α and prevented colon tissue injury to some extent. Notably, the results for the SD11 control mice were identical to those for the normal control mice. Overall, our findings that SD11 delays liver deterioration and reduces colon lesions in diabetic mice provide evidence for the use of SD11 as an effective strategy to improve diabetes-related symptoms.
Collapse
Affiliation(s)
- Waraporn Ruathong
- Faculty of Science, Division of Health and Applied Sciences, Prince of Songkla University, Songkhla, Thailand
| | - Pissared Khuituan
- Faculty of Science, Division of Health and Applied Sciences, Prince of Songkla University, Songkhla, Thailand
| | - Saranya Peerakietkhajorn
- Faculty of Science, Division of Biological Science, Prince of Songkla University, Songkhla, Thailand
| | - Rawee Teanpaisan
- Faculty of Dentistry, Research Center of Excellence for Oral Health, Prince of Songkla University, Hat Yai, Thailand
| | - Jongdee Nopparat
- Faculty of Science, Division of Health and Applied Sciences, Prince of Songkla University, Songkhla, Thailand
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| |
Collapse
|
26
|
Lee J, An H, Kim CS, Lee S. The methyltransferase MLL4 promotes nonalcoholic steatohepatitis by enhancing NF-κB signaling. J Biol Chem 2024; 300:107984. [PMID: 39542242 DOI: 10.1016/j.jbc.2024.107984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/28/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a growing health problem worldwide, ranging from non-alcoholic fatty liver (NAFL) to the more severe metabolic non-alcoholic steatohepatitis (NASH). Although many studies have elucidated the pathogenesis of NAFLD, the epigenetic regulatory mechanism from NAFL to NASH remains incompletely understood. The histone H3 lysine 4 methyltransferase, MLL4 (also called KMT2D), is a critical epigenetic transcriptional coactivator that mediates overnutrition-induced steatosis in mice, but its potential role in the progression of NASH remains largely unknown. Here, we show that mice lacking the one allele of the Mll4 gene are resistant to hepatic steatosis, inflammation, and fibrosis in NASH conditions compared to wild-type controls. Transcriptome analysis of the livers of control and Mll4+/- mice identified pro-inflammatory genes regulated by the nuclear factor kappa B (NF-κB) signaling pathway as major target genes of MLL4. We show that MLL4 binds to p65 and that MLL4 is required for NF-κB transactivation. Myeloid-specific Mll4 knockout mice showed an almost complete block of NASH, while hepatocyte-specific Mll4 knockout mice showed mild inhibition of steatosis. Pro-inflammatory M1 polarization is decreased and anti-inflammatory M2 polarization is increased in liver macrophages from myeloid-specific Mll4 knockout mice. Importantly, we show that histone H3-lysine 4 methylation mediated by the MLL4-complex plays a critical role in promoting the expression of Ccl2 in hepatocytes and M1 marker genes in macrophages. Our results demonstrate that MLL4, through the NF-κB-MLL4 regulatory axis, exacerbates steatohepatitis in the context of an inflammatory response and represents a potential therapeutic target for NASH.
Collapse
Affiliation(s)
- Junekyoung Lee
- Research Institute of Pharmaceutical Sciences, Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Hyejin An
- Research Institute of Pharmaceutical Sciences, Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Chong-Su Kim
- Department of Food and Nutrition, College of Natural Information Sciences, Dongduk Women's University, Seoul, South Korea
| | - Seunghee Lee
- Research Institute of Pharmaceutical Sciences, Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, South Korea.
| |
Collapse
|
27
|
Bai Y, Song Y, Li M, Ou J, Hu H, Xu N, Cao M, Wang S, Chen L, Cheng G, Li Z, Liu G, Wang J, Zhang W, Yang C. Dissection of molecular mechanisms of liver injury induced by microcystin-leucine arginine via single-cell RNA-sequencing. J Environ Sci (China) 2024; 145:164-179. [PMID: 38844317 DOI: 10.1016/j.jes.2023.08.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 07/28/2024]
Abstract
The occurrence of poisoning incidents caused by cyanobacterial blooms has aroused wide public concern. Microcystin-leucine arginine (MC-LR) is a well-established toxin produced by cyanobacterial blooms, which is widely distributed in eutrophic waters. MC-LR is not only hazardous to the water environment but also exerts multiple toxic effects including liver toxicity in both humans and animals. However, the underlying mechanisms of MC-LR-induced liver toxicity are unclear. Herein, we used advanced single-cell RNA sequencing technology to characterize MC-LR-induced liver injury in mice. We established the first single-cell atlas of mouse livers in response to MC-LR. Our results showed that the differentially expressed genes and pathways in diverse cell types of liver tissues of mice treated with MC-LR are highly heterogeneous. Deep analysis showed that MC-LR induced an increase in a subpopulation of hepatocytes that highly express Gstm3, which potentially contributed to hepatocyte apoptosis in response to MC-LR. Moreover, MC-LR increased the proportion and multiple subtypes of Kupffer cells with M1 phenotypes and highly expressed proinflammatory genes. Furthermore, the MC-LR increased several subtypes of CD8+ T cells with highly expressed multiple cytokines and chemokines. Overall, apart from directly inducing hepatocytes apoptosis, MC-LR activated proinflammatory Kupffer cell and CD8+ T cells, and their interaction may constitute a hostile microenvironment that contributes to liver injury. Our findings not only present novel insight into underlying molecular mechanisms but also provide a valuable resource and foundation for additional discovery of MC-LR-induced liver toxicity.
Collapse
Affiliation(s)
- Yunmeng Bai
- Dongguan Maternal and Child Health Care Hospital, Postdoctoral Innovation Practice Base of Southern Medical University, Dongguan 523125, China; Division of Thyroid and Breast Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Yali Song
- Dongguan Maternal and Child Health Care Hospital, Postdoctoral Innovation Practice Base of Southern Medical University, Dongguan 523125, China
| | - Miaoran Li
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jinhuan Ou
- Division of Thyroid and Breast Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Hong Hu
- Division of Thyroid and Breast Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Nan Xu
- Division of Thyroid and Breast Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Min Cao
- Division of Thyroid and Breast Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Siyu Wang
- Faculty of Brain Sciences, University College London, WC1E 6BT, UK
| | - Lin Chen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Guangqing Cheng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zhijie Li
- Division of Thyroid and Breast Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Gang Liu
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Jigang Wang
- Dongguan Maternal and Child Health Care Hospital, Postdoctoral Innovation Practice Base of Southern Medical University, Dongguan 523125, China; Division of Thyroid and Breast Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Wei Zhang
- Division of Thyroid and Breast Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China.
| | - Chuanbin Yang
- Division of Thyroid and Breast Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China.
| |
Collapse
|
28
|
Khanna K, Eul E, Yan H, Faccio R. Delayed macrophage targeting by clodronate liposomes worsens the progression of cytokine storm syndrome. Front Immunol 2024; 15:1477449. [PMID: 39530102 PMCID: PMC11550973 DOI: 10.3389/fimmu.2024.1477449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Excessive macrophage activation and production of pro-inflammatory cytokines are hallmarks of the Cytokine Storm Syndrome (CSS), a lethal condition triggered by sepsis, autoimmune disorders, and cancer immunotherapies. While depletion of macrophages at disease onset protects from lethality in an infection-induced CSS murine model, patients are rarely diagnosed early, hence the need to characterize macrophage populations during CSS progression and assess the therapeutic implications of macrophage targeting after disease onset. In this study, we identified MHCII+F4/80+Tim4- macrophages as the primary contributors to the pro-inflammatory environment in CSS, while CD206+F4/80+Tim4+ macrophages, with an anti-inflammatory profile, become outnumbered. Additionally, we observed an expansion of Tim4- macrophages coinciding with increased hematopoietic stem progenitor cells and reduction of committed myeloid progenitors in bone marrow and spleen. Critically, macrophage targeting with clodronate liposomes at disease onset prolonged survival, while their targeting in mice with established CSS exacerbated disease severity, leading to a more dramatic loss of Tim4+ macrophages and an imbalance in pro- versus anti-inflammatory Tim4- macrophage ratio. Our findings highlight the significance of timing in macrophage-targeted interventions for effective management of CSS and suggest potential therapeutic strategies for diseases characterized by uncontrolled inflammation, such as sepsis.
Collapse
Affiliation(s)
- Kunjan Khanna
- Department of Orthopedics, Washington University in St Louis, St Louis, MO, United States
| | - Emily Eul
- Department of Orthopedics, Washington University in St Louis, St Louis, MO, United States
| | - Hui Yan
- Department of Orthopedics, Washington University in St Louis, St Louis, MO, United States
| | - Roberta Faccio
- Department of Orthopedics, Washington University in St Louis, St Louis, MO, United States
- Shriners Hospital for Children, St Louis, MO, United States
| |
Collapse
|
29
|
Yuan H, Li Y, Kong Z, Peng L, Song J, Hou X, Zhang W, Liu R, Feng T, Zhu C. IL-33-Pretreated Mesenchymal Stem Cells Attenuate Acute Liver Failure by Improving Homing and Polarizing M2 Macrophages. Stem Cells Int 2024; 2024:1273099. [PMID: 39478979 PMCID: PMC11524710 DOI: 10.1155/2024/1273099] [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: 01/20/2024] [Revised: 07/05/2024] [Accepted: 09/18/2024] [Indexed: 11/02/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are highly effective in the treatment of acute liver failure (ALF). The efficacy of MSCs is closely related to the inflammatory environment. Therefore, we investigated the functional changes of MSCs in response to interleukin-33 (IL-33) stimulation. The results showed that bone marrow mesenchymal stem cells (BMSCs) pretreated with IL-33 had increased CCR2 expression, targeted CCL2 in the injured liver tissue, and improved the migration ability. Under LPS stimulation, the NF-κB pathway of BMDM was activated, and its phenotype polarized to the M1-type, while BMSCs pretreated with IL-33 inhibited the NF-κB pathway and enhanced M2 macrophage polarization. The M2-type macrophages could further inhibit hepatocytes inflammation, reduce hepatocytes apoptosis, and promote hepatocytes repair. These results suggest that IL-33 can enhance the efficacy of BMSCs in ALF and provide a new strategy for cell therapy of liver diseases.
Collapse
Affiliation(s)
- Hui Yuan
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuwen Li
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zihao Kong
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Linya Peng
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiali Song
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoxue Hou
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wen Zhang
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Liu
- Department of Infectious and Tropical Diseases, The Second Affiliated Hospital, NHC Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, China
| | - Tiantong Feng
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chuanlong Zhu
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Infectious and Tropical Diseases, The Second Affiliated Hospital, NHC Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, China
| |
Collapse
|
30
|
Graham CT, Gordon S, Kubes P. A historical perspective of Kupffer cells in the context of infection. Cell Tissue Res 2024:10.1007/s00441-024-03924-4. [PMID: 39392500 DOI: 10.1007/s00441-024-03924-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
The Kupffer cell was first discovered by Karl Wilhelm von Kupffer in 1876, labeling them as "Sternzellen." Since their discovery as the primary macrophages of the liver, researchers have gradually gained an in-depth understanding of the identity, functions, and influential role of Kupffer cells, particularly in infection. It is becoming clear that Kupffer cells perform important tissue-specific functions in homeostasis and disease. Stationary in the sinusoids of the liver, Kupffer cells have a high phagocytic capacity and are adept in clearing the bloodstream of foreign material, toxins, and pathogens. Thus, they are indispensable to host defense and prevent the dissemination of bacteria during infections. To highlight the importance of this cell, this review will explore the history of the Kupffer cell in the context of infection beginning with its discovery to the present day.
Collapse
Affiliation(s)
- Carolyn T Graham
- Department of Physiology and Pharmacology, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
| | - Siamon Gordon
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wenhua 1st Road Guishan Dist., Taoyuan, Taiwan
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| |
Collapse
|
31
|
Brüggemann Y, Klöhn M, Wedemeyer H, Steinmann E. Hepatitis E virus: from innate sensing to adaptive immune responses. Nat Rev Gastroenterol Hepatol 2024; 21:710-725. [PMID: 39039260 DOI: 10.1038/s41575-024-00950-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/29/2024] [Indexed: 07/24/2024]
Abstract
Hepatitis E virus (HEV) infections are a major cause of acute viral hepatitis in humans worldwide. In immunocompetent individuals, the majority of HEV infections remain asymptomatic and lead to spontaneous clearance of the virus, and only a minority of individuals with infection (5-16%) experience symptoms of acute viral hepatitis. However, HEV infections can cause up to 30% mortality in pregnant women, become chronic in immunocompromised patients and cause extrahepatic manifestations. A growing body of evidence suggests that the host immune response to infection with different HEV genotypes is a critical determinant of distinct HEV infection outcomes. In this Review, we summarize key components of the innate and adaptive immune responses to HEV, including the underlying immunological mechanisms of HEV associated with acute and chronic liver failure and interactions between T cell and B cell responses. In addition, we discuss the current status of vaccines against HEV and raise outstanding questions regarding the immune responses induced by HEV and treatment of the disease, highlighting areas for future investigation.
Collapse
Affiliation(s)
- Yannick Brüggemann
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Mara Klöhn
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner Sites Hannover-Braunschweig, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Eike Steinmann
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany.
- German Center for Infection Research (DZIF), External Partner Site, Bochum, Germany.
| |
Collapse
|
32
|
Yan J, Nie Y, Chen X, Ding M, Zhang S. Mechanistic study of fructus aurantii (Quzhou origin) in regulating ileal reg3g in the treatment for NASH. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155924. [PMID: 39098169 DOI: 10.1016/j.phymed.2024.155924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 07/06/2024] [Accepted: 07/27/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) is a critical stage in the progression of non-alcoholic fatty liver disease (NAFLD), characterized by obvious inflammation and fibrosis. Because of its high incidence rate and serious consequences, NASH is becoming a global health problem. The influence of endotoxin translocation on NASH is receiving attention. As a traditional Chinese herb that effectively improves hepatic inflammation, Fructus Aurantii (Quzhou origin, FAQ) is widely used in the clinical treatment of NASH. However, the intervention mechanism of FAQ on reg3g and related endotoxin translocation remains unclear. AIM To study the mechanism of the impact by which ileal regenerating family member 3 gamma (reg3g) deficiency and subsequent endotoxin translocation impact the progression of NASH; To elucidate the efficacy and mechanism of FAQ in the treatment of NASH. METHODS Clinical serum, ileal tissue, and dynamic NASH model-related analyses collectively confirmed that reg3g is a pivotal gene associated with NASH. Reg3g-/- mice were used to assess the impact of reg3g on liver injury, inflammation, and fibrosis, as well as the underlying mechanism involved. In vitro studies elucidated the regulatory effects of FAQ on reg3g, intestinal barrier function, and intestinal permeability. Subsequently, the efficacy of FAQ was investigated in NASH mouse models. Pathological examinations combined with Western blotting (WB), immunohistochemistry (IHC), and multiplex immunohistochemical (mIHC) analyses were used to evaluate the effects of FAQ on mucosal repair and barrier function. Transepithelial electrical resistance (TEER), fluorescein isothiocyanate-dextran 4 (FD-4) experiments, coupled with enzyme linked immunosorbent assay (ELISA) and chromogenic LAL endotoxin assay were used to confirm intestinal permeability and endotoxin translocation. The results of WB and mIHC reflected the levels of endotoxin recruitment and M1 macrophage polarization in the liver. Parameters such as body weight, transaminases, and cholesterol were utilized to assess the metabolic effects of FAQ. RESULTS Decreased expression of reg3g was associated with the progression of NASH. Ileal deficiency in reg3g resulted in damage to the intestinal barrier and permeability, leading to the recruitment of endotoxins via the 'gut-liver' axis to the liver, causing the polarization of M1 macrophages, release of inflammatory factors, excessive inflammation, and activation of hepatic stellate cells (HSCs), leading to fibrosis. FAQ significantly upregulated ileal reg3g expression and the expression of intestinal barrier-related proteins tight junction protein 1 (ZO-1) and occludin (OLCN) in mice (p < 0.05), thereby improving intestinal barrier function and permeability. Reduced intestinal permeability led to decreases in endotoxins entering the bloodstream and accumulating in the liver (p < 0.05). The expression of CD68 suggested reduced polarization of M1 macrophages. Expression levels of actin alpha 2, smooth muscle actin (α-SMA) and extracellular matrix (ECM)-related proteins also decreased, indicating improved liver fibrosis. CONCLUSION FAQ ameliorates NASH by upregulating the expression of reg3g. The upregulation of reg3g contributes to the repair of the intestinal barrier and permeability, reducing the recruitment of endotoxins and subsequent polarization of M1 macrophages, excessive inflammation, and fibrosis.
Collapse
Affiliation(s)
- Junbin Yan
- The Second Affiliated Hospital of Zhejiang Chinese Medical University (The Xin Hua Hospital of Zhejiang Province), Hangzhou 310000, China
| | - Yunmeng Nie
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Xinli Chen
- The Second Affiliated Hospital of Zhejiang Chinese Medical University (The Xin Hua Hospital of Zhejiang Province), Hangzhou 310000, China
| | - Menglu Ding
- The Second Affiliated Hospital of Zhejiang Chinese Medical University (The Xin Hua Hospital of Zhejiang Province), Hangzhou 310000, China
| | - Shuo Zhang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University (The Xin Hua Hospital of Zhejiang Province), Hangzhou 310000, China; Key Laboratory of Traditional Chinese Medicine for the treatment of Intestine-Liver of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
33
|
Overstreet AMC, Burge M, Bellar A, McMullen M, Czarnecki D, Huang E, Pathak V, Finney C, Vij R, Dasarathy S, Dasarathy J, Streem D, Welch N, Rotroff D, Schmitt AM, Nagy LE, Messer JS. Evidence that extracellular HSPB1 contributes to inflammation in alcohol-associated hepatitis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.06.24313193. [PMID: 39281760 PMCID: PMC11398598 DOI: 10.1101/2024.09.06.24313193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Background and aims Alcohol-associated hepatitis (AH) is the most life-threatening form of alcohol-associated liver disease (ALD). AH is characterized by severe inflammation attributed to increased levels of ethanol, microbes or microbial components, and damage-associated molecular pattern (DAMP) molecules in the liver. HSPB1 (Heat Shock Protein Family B (Small) Member 1; also known as Hsp25/27) is a DAMP that is rapidly increased in and released from cells experiencing stress, including hepatocytes. The goal of this study was to define the role of HSPB1 in AH pathophysiology. Methods Serum HSPB1 was measured in a retrospective study of 184 heathy controls (HC), heavy alcohol consumers (HA), patients with alcohol-associated cirrhosis (AC), and patients with AH recruited from major hospital centers. HSPB1 was also retrospectively evaluated in liver tissue from 10 HC and AH patients and an existing liver RNA-seq dataset. Finally, HSPB1 was investigated in a murine Lieber-DeCarli diet model of early ALD as well as cellular models of ethanol stress in hepatocytes and hepatocyte-macrophage communication during ethanol stress. Results Circulating HSPB1 was significantly increased in AH patients and levels positively correlated with disease-severity scores. Likewise, HSPB1 was increased in the liver of patients with severe AH and in the liver of ethanol-fed mice. In vitro , ethanol-stressed hepatocytes released HSPB1, which then triggered TNFα-mediated inflammation in macrophages. Anti-HSPB1 antibody prevented TNFα release from macrophages exposed to media conditioned by ethanol-stressed hepatocytes. Conclusions Our findings support investigation of HSPB1 as both a biomarker and therapeutic target in ALD. Furthermore, this work demonstrates that anti-HSPB1 antibody is a rational approach to targeting HSPB1 with the potential to block inflammation and protect hepatocytes, without inactivating host defense. GRAPHICAL ABSTRACT HIGHLIGHTS HSPB1 is significantly increased in serum and liver of patients with alcohol-associated hepatitis.Ethanol consumption leads to early increases in HSPB1 in the mouse liver.Hepatocytes subjected to ethanol stress release HSPB1 into the extracellular environment where it activates TNFα-mediated inflammation in macrophages.Anti-HSPB1 antibody blocks hepatocyte-triggered TNFα in a model of hepatocyte-macrophage communication during ethanol stress.
Collapse
|
34
|
Chen S, Zeng Q, Cai X, Xue J, Yin G, Song P, Tang L, Klein C, Tacke F, Guillot A, Liu H. Multiomics analyses decipher intricate changes in the cellular and metabolic landscape of steatotic livers upon dietary restriction and sleeve gastrectomy. Int J Biol Sci 2024; 20:4438-4457. [PMID: 39247824 PMCID: PMC11380448 DOI: 10.7150/ijbs.98362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 08/04/2024] [Indexed: 09/10/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a chronic, progressive liver disease that encompasses a spectrum of steatosis, steatohepatitis (or MASH), and fibrosis. Evidence suggests that dietary restriction (DR) and sleeve gastrectomy (SG) can lead to remission of hepatic steatosis and inflammation through weight loss, but it is unclear whether these procedures induce distinct metabolic or immunological changes in MASLD livers. This study aims to elucidate the intricate hepatic changes following DR, SG or sham surgery in rats fed a high-fat diet as a model of obesity-related MASLD, in comparison to a clinical cohort of patients undergoing SG. Single-cell and single-nuclei transcriptome analysis, spatial metabolomics, and immunohistochemistry revealed the liver landscape, while circulating biomarkers were measured in serum samples. Artificial intelligence (AI)-assisted image analysis characterized the spatial distribution of hepatocytes, myeloid cells and lymphocytes. In patients and experimental MASLD rats, SG improved body mass index, circulating liver injury biomarkers and triglyceride levels. Both DR and SG attenuated liver steatosis and fibrosis in rats. Metabolism-related genes (Ppara, Cyp2e1 and Cyp7a1) were upregulated in hepatocytes upon DR and SG, while SG broadly upregulated lipid metabolism on cholangiocytes, monocytes, macrophages, and neutrophils. Furthermore, SG promoted restorative myeloid cell accumulation in the liver not only ameliorating inflammation but activating liver repair processes. Regions with potent myeloid infiltration were marked with enhanced metabolic capacities upon SG. Additionally, a disruption of periportal hepatocyte functions was observed upon DR. In conclusion, this study indicates a dynamic cellular crosstalk in steatotic livers of patients undergoing SG. Notably, PPARα- and gut-liver axis-related processes, and metabolically active myeloid cell infiltration indicate intervention-related mechanisms supporting the indication of SG for the treatment of MASLD.
Collapse
Affiliation(s)
- Shuai Chen
- Department of General Surgery, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Qinghe Zeng
- Laboratoire d'Informatique Paris Descartes (LIPADE), Université Paris Cité, Paris 75014, France
- Centre d'Histologie, d'Imagerie et de Cytométrie (CHIC), Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris 75014, France
| | | | - Jiaming Xue
- Department of General Surgery, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Guo Yin
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin 13353, Germany
| | - Peng Song
- Department of General Surgery, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Liming Tang
- Department of General Surgery, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Christophe Klein
- Centre d'Histologie, d'Imagerie et de Cytométrie (CHIC), Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris 75014, France
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin 13353, Germany
| | - Adrien Guillot
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin 13353, Germany
| | - Hanyang Liu
- Department of General Surgery, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin 13353, Germany
| |
Collapse
|
35
|
Streutker EM, Devamoglu U, Vonk MC, Verdurmen WPR, Le Gac S. Fibrosis-on-Chip: A Guide to Recapitulate the Essential Features of Fibrotic Disease. Adv Healthc Mater 2024; 13:e2303991. [PMID: 38536053 DOI: 10.1002/adhm.202303991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/15/2024] [Indexed: 05/05/2024]
Abstract
Fibrosis, which is primarily marked by excessive extracellular matrix (ECM) deposition, is a pathophysiological process associated with many disorders, which ultimately leads to organ dysfunction and poor patient outcomes. Despite the high prevalence of fibrosis, currently there exist few therapeutic options, and importantly, there is a paucity of in vitro models to accurately study fibrosis. This review discusses the multifaceted nature of fibrosis from the viewpoint of developing organ-on-chip (OoC) disease models, focusing on five key features: the ECM component, inflammation, mechanical cues, hypoxia, and vascularization. The potential of OoC technology is explored for better modeling these features in the context of studying fibrotic diseases and the interplay between various key features is emphasized. This paper reviews how organ-specific fibrotic diseases are modeled in OoC platforms, which elements are included in these existing models, and the avenues for novel research directions are highlighted. Finally, this review concludes with a perspective on how to address the current gap with respect to the inclusion of multiple features to yield more sophisticated and relevant models of fibrotic diseases in an OoC format.
Collapse
Affiliation(s)
- Emma M Streutker
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Utku Devamoglu
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnoloygy and TechMed Centre, Organ-on-Chip Centre, University of Twente, Drienerlolaan 5, Enschede, 7522 NB, The Netherlands
| | - Madelon C Vonk
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Wouter P R Verdurmen
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Séverine Le Gac
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnoloygy and TechMed Centre, Organ-on-Chip Centre, University of Twente, Drienerlolaan 5, Enschede, 7522 NB, The Netherlands
| |
Collapse
|
36
|
Chen C, Feng D, Wang Y, Yao T, Mackowiak B, Gao B. Necrotic Liver Lesion Resolution: Another Mode of Liver Regeneration. Semin Liver Dis 2024; 44:333-342. [PMID: 38955211 DOI: 10.1055/a-2358-9505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
The liver has the great ability to regenerate after partial resection or injury, and the mechanisms underlying liver regeneration have been extensively investigated. Interestingly, acute liver injuries triggered by various etiologies are associated with the formation of necrotic lesions, and such necrotic lesions are also rapidly resolved. However, how necrotic liver lesions are repaired has not been carefully investigated until recently. In this review, we briefly summarize the spatiotemporal process of necrotic liver lesion resolution in several liver injury models including immune-mediated liver injury and drug-induced liver injury. The roles of liver nonparenchymal cells and infiltrating immune cells in controlling necrotic liver lesion resolution are discussed, which may help identify potential therapies for acute liver injury and failure.
Collapse
Affiliation(s)
- Cheng Chen
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Yang Wang
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Tiantian Yao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Bryan Mackowiak
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
37
|
Wang J, Lu H, Li Q. Hepatic macrophage niche: a bridge between HBV-mediated metabolic changes with intrahepatic inflammation. Front Immunol 2024; 15:1414594. [PMID: 39091506 PMCID: PMC11291371 DOI: 10.3389/fimmu.2024.1414594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024] Open
Abstract
Hepatitis B Virus (HBV) is a stealthy and insidious pathogen capable of inducing chronic necro-inflammatory liver disease and hepatocellular carcinoma (HCC), resulting in over one million deaths worldwide per year. The traditional understanding of Chronic Hepatitis B (CHB) progression has focused on the complex interplay among ongoing virus replication, aberrant immune responses, and liver pathogenesis. However, the dynamic progression and crucial factors involved in the transition from HBV infection to immune activation and intrahepatic inflammation remain elusive. Recent insights have illuminated HBV's exploitation of the sodium taurocholate co-transporting polypeptide (NTCP) and manipulation of the cholesterol transport system shared between macrophages and hepatocytes for viral entry. These discoveries deepen our understanding of HBV as a virus that hijacks hepatocyte metabolism. Moreover, hepatic niche macrophages exhibit significant phenotypic and functional diversity, zonal characteristics, and play essential roles, either in maintaining liver homeostasis or contributing to the pathogenesis of chronic liver diseases. Therefore, we underscore recent revelations concerning the importance of hepatic niche macrophages in the context of viral hepatitis. This review particularly emphasizes the significant role of HBV-induced metabolic changes in hepatic macrophages as a key factor in the transition from viral infection to immune activation, ultimately culminating in liver inflammation. These metabolic alterations in hepatic macrophages offer promising targets for therapeutic interventions and serve as valuable early warning indicators, shedding light on the disease progression.
Collapse
Affiliation(s)
- Jun Wang
- The Third People’s Hospital of Shenzhen (National Clinical Research Center for Infectious Diseases) and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
- Clinical Research Center, The Fifth People’s Hospital of Wuxi, Jiangnan University, Wuxi, Jiangsu, China
| | - Hongzhou Lu
- The Third People’s Hospital of Shenzhen (National Clinical Research Center for Infectious Diseases) and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Qian Li
- The Third People’s Hospital of Shenzhen (National Clinical Research Center for Infectious Diseases) and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
38
|
Zeng Y, Wu Z, Chen G, Liu G, Zhang B, Zhou Y, Chen M, Yao R, Shi Y. Peripheral Injection of hUC-MSCs in the Treatment of Acute Liver Failure: A Pre-Clinical Cohort Study in Rhesus Monkeys. Stem Cells Int 2024; 2024:4654912. [PMID: 39045027 PMCID: PMC11265939 DOI: 10.1155/2024/4654912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 06/19/2024] [Accepted: 06/27/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Using a toxin-induced lethal acute liver failure (ALF) monkey model, we have recently shown that early peripheral infusion of human umbilical cord mesenchymal stem cells (hUC-MSCs) can alleviate liver damage and improve animal survival by suppressing the activation of circulating monocytes and the subsequent cytokine storm. Here, we explored whether the administration of hUC-MSCs could still improve ALF when the cytokine storm is fully developed. METHOD We treated ALF monkeys with peripheral delivery of hUC-MSCs at 48 hr after toxin challenge. Liver indices, histology, imaging, and animal survival were recorded and analyzed. RESULTS In our cohort study, we conducted and demonstrated that the infusion of hUC-MSCs significantly improved liver histology, effectively controlled inflammatory cytokine storms, and increased survival rates. Additionally, the administration of a higher dose of hUC-MSCs (2 × 107/monkey) yielded superior outcomes compared to a lower dose (1 × 107/monkey). CONCLUSION Treatment of hUC-MSCs can significantly improve the pathological and survival outcomes of ALF even when the cytokine storm has been fully developed, indicating a promising clinical solution for ALF.
Collapse
Affiliation(s)
- Yuting Zeng
- Liver Transplant CenterTransplant Center and Key Laboratory of Transplant Engineering and ImmunologyNHCWest China HospitalSichuan University, Chengdu, China
- Institute of clinical PathologyWest China HospitalSichuan University, Chengdu, China
| | - Zhenru Wu
- Liver Transplant CenterTransplant Center and Key Laboratory of Transplant Engineering and ImmunologyNHCWest China HospitalSichuan University, Chengdu, China
- Institute of clinical PathologyWest China HospitalSichuan University, Chengdu, China
| | - Gen Chen
- Development and Application of Human Major Disease Monkey Model Key Laboratory of SichuanSichuan Yibin Horizontal and Vertical Biotechnology Co., Ltd., Yibin 644601, China
| | - Guoqiang Liu
- Development and Application of Human Major Disease Monkey Model Key Laboratory of SichuanSichuan Yibin Horizontal and Vertical Biotechnology Co., Ltd., Yibin 644601, China
| | - Bo Zhang
- Sichuan Stem Cell Bank and Sichuan Neo-Life Stem Cell Biotech Inc., Chengdu 610037, China
| | - Yongjie Zhou
- Laboratory of Liver TransplantationWest China HospitalSichuan University, Chengdu 610041, China
| | - Menglin Chen
- Liver Transplant CenterTransplant Center and Key Laboratory of Transplant Engineering and ImmunologyNHCWest China HospitalSichuan University, Chengdu, China
- Institute of clinical PathologyWest China HospitalSichuan University, Chengdu, China
| | - Rong Yao
- Department of Emergency MedicineEmergency Medical LaboratoryWest China HospitalSichuan University, Chengdu, Sichuan, China
| | - Yujun Shi
- Liver Transplant CenterTransplant Center and Key Laboratory of Transplant Engineering and ImmunologyNHCWest China HospitalSichuan University, Chengdu, China
- Institute of clinical PathologyWest China HospitalSichuan University, Chengdu, China
| |
Collapse
|
39
|
Tao Y, Wang Y, Wang M, Tang H, Chen E. Mesenchymal Stem Cells Alleviate Acute Liver Failure through Regulating Hepatocyte Apoptosis and Macrophage Polarization. J Clin Transl Hepatol 2024; 12:571-580. [PMID: 38974955 PMCID: PMC11224903 DOI: 10.14218/jcth.2023.00557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/06/2024] [Accepted: 03/18/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND AND AIMS Acute liver failure (ALF) is a life-threatening clinical problem with limited treatment options. Administration of human umbilical cord mesenchymal stem cells (hUC-MSCs) may be a promising approach for ALF. This study aimed to explore the role of hUC-MSCs in the treatment of ALF and the underlying mechanisms. METHODS A mouse model of ALF was induced by lipopolysaccharide and d-galactosamine administration. The therapeutic effects of hUC-MSCs were evaluated by assessing serum enzyme activity, histological appearance, and cell apoptosis in liver tissues. The apoptosis rate was analyzed in AML12 cells. The levels of inflammatory cytokines and the phenotype of RAW264.7 cells co-cultured with hUC-MSCs were detected. The C-Jun N-terminal kinase/nuclear factor-kappa B signaling pathway was studied. RESULTS The hUC-MSCs treatment decreased the levels of serum alanine aminotransferase and aspartate aminotransferase, reduced pathological damage, alleviated hepatocyte apoptosis, and reduced mortality in vivo. The hUC-MSCs co-culture reduced the apoptosis rate of AML12 cells in vitro. Moreover, lipopolysaccharide-stimulated RAW264.7 cells had higher levels of tumor necrosis factor-α, interleukin-6, and interleukin-1β and showed more CD86-positive cells, whereas the hUC-MSCs co-culture reduced the levels of the three inflammatory cytokines and increased the ratio of CD206-positive cells. The hUC-MSCs treatment inhibited the activation of phosphorylated (p)-C-Jun N-terminal kinase and p-nuclear factor-kappa B not only in liver tissues but also in AML12 and RAW264.7 cells co-cultured with hUC-MSCs. CONCLUSIONS hUC-MSCs could alleviate ALF by regulating hepatocyte apoptosis and macrophage polarization, thus hUC-MSC-based cell therapy may be an alternative option for patients with ALF.
Collapse
Affiliation(s)
- Yachao Tao
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Yonghong Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Menglan Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Enqiang Chen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
40
|
An R, Blackwell VK, Harandi B, Gibbons AC, Siu O, Irby I, Rees A, Cornejal N, Sattler KM, Sheng T, Syracuse NC, Loftus D, Santa Maria SR, Cekanaviciute E, Reinsch SS, Ray HE, Paul AM. Influence of the spaceflight environment on macrophage lineages. NPJ Microgravity 2024; 10:63. [PMID: 38862517 PMCID: PMC11166655 DOI: 10.1038/s41526-023-00293-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 05/25/2023] [Indexed: 06/13/2024] Open
Abstract
Spaceflight and terrestrial spaceflight analogs can alter immune phenotypes. Macrophages are important immune cells that bridge the innate and adaptive immune systems and participate in immunoregulatory processes of homeostasis. Furthermore, macrophages are critically involved in initiating immunity, defending against injury and infection, and are also involved in immune resolution and wound healing. Heterogeneous populations of macrophage-type cells reside in many tissues and cause a variety of tissue-specific effects through direct or indirect interactions with other physiological systems, including the nervous and endocrine systems. It is vital to understand how macrophages respond to the unique environment of space to safeguard crew members with appropriate countermeasures for future missions in low Earth orbit and beyond. This review highlights current literature on macrophage responses to spaceflight and spaceflight analogs.
Collapse
Affiliation(s)
- Rocky An
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Cornell University, Department of Biological and Environmental Engineering and Sibley School of Mechanical and Aerospace Engineering, Ithaca, NY, 14853, USA
| | - Virginia Katherine Blackwell
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Massachusetts Institute of Technology, Department of Biology, Cambridge, MA, 02139, USA
| | - Bijan Harandi
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Tufts University, Department of Chemistry, Medford, MA, 02155, USA
| | - Alicia C Gibbons
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- University of California San Diego, Department of Cellular and Molecular Medicine, La Jolla, CA, 92093, USA
| | - Olivia Siu
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Embry-Riddle Aeronautical University, Department of Human Factors and Behavioral Neurobiology, Daytona Beach, FL, 32114, USA
| | - Iris Irby
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Amy Rees
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Nadjet Cornejal
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Brooklyn College, Department of Natural and Behavioral Sciences, Brooklyn, NY, 11210, USA
| | - Kristina M Sattler
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Ohio State University, Department of Physiology and Cell Biology, Columbus, OH, 43210, USA
| | - Tao Sheng
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- University of Pittsburgh, Department of Computer Science, Pittsburgh, PA, 15260, USA
| | - Nicholas C Syracuse
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- North Carolina State University, Department of Molecular and Structural Biochemistry and Department of Biological Sciences, Raleigh, NC, 27695, USA
| | - David Loftus
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA, 94035, USA
| | - Sergio R Santa Maria
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA, 94035, USA
| | - Egle Cekanaviciute
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA, 94035, USA
| | - Sigrid S Reinsch
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA, 94035, USA
| | - Hami E Ray
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA, 94035, USA
- ASRC Federal, Inc, Beltsville, MD, 20705, USA
| | - Amber M Paul
- Embry-Riddle Aeronautical University, Department of Human Factors and Behavioral Neurobiology, Daytona Beach, FL, 32114, USA.
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA, 94035, USA.
- Blue Marble Space Institute of Science, Seattle, WA, 98104, USA.
| |
Collapse
|
41
|
Holt M, Lin J, Cicka M, Wong A, Epelman S, Lavine KJ. Dissecting and Visualizing the Functional Diversity of Cardiac Macrophages. Circ Res 2024; 134:1791-1807. [PMID: 38843293 DOI: 10.1161/circresaha.124.323817] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024]
Abstract
Cardiac macrophages represent a functionally diverse population of cells involved in cardiac homeostasis, repair, and remodeling. With recent advancements in single-cell technologies, it is possible to elucidate specific macrophage subsets based on transcriptional signatures and cell surface protein expression to gain a deep understanding of macrophage diversity in the heart. The use of fate-mapping technologies and parabiosis studies have provided insight into the ontogeny and dynamics of macrophages identifying subsets derived from embryonic and adult definitive hematopoietic progenitors that include tissue-resident and bone marrow monocyte-derived macrophages, respectively. Within the heart, these subsets have distinct tissue niches and functional roles in the setting of homeostasis and disease, with cardiac resident macrophages representing a protective cell population while bone marrow monocyte-derived cardiac macrophages have a context-dependent effect, triggering both proinflammatory tissue injury, but also promoting reparative functions. With the increased understanding of the clinical relevance of cardiac macrophage subsets, there has been an increasing need to detect and measure cardiac macrophage compositions in living animals and patients. New molecular tracers compatible with positron emission tomography/computerized tomography and positron emission tomography/ magnetic resonance imaging have enabled investigators to noninvasively and serially visualize cardiac macrophage subsets within the heart to define associations with disease and measure treatment responses. Today, advancements within this thriving field are poised to fuel an era of clinical translation.
Collapse
Affiliation(s)
- Megan Holt
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine (M.H., M.C., K.J.L.)
| | - Julia Lin
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (J.L., A.W., S.E.)
- Department of Immunology, University of Toronto, ON, Canada (J.L., A.W., S.E.)
| | - Markus Cicka
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine (M.H., M.C., K.J.L.)
| | - Anthony Wong
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (J.L., A.W., S.E.)
- Department of Immunology, University of Toronto, ON, Canada (J.L., A.W., S.E.)
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (J.L., A.W., S.E.)
- Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada (S.E.)
- Department of Immunology, University of Toronto, ON, Canada (J.L., A.W., S.E.)
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada (S.E.)
| | - Kory J Lavine
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine (M.H., M.C., K.J.L.)
| |
Collapse
|
42
|
Abstract
Acute liver injury (ALI), that is, the development of reduced liver function in patients without preexisting liver disease, can result from a wide range of causes, such as viral or bacterial infection, autoimmune disease, or adverse reaction to prescription and over-the-counter medications. ALI patients present with a complex coagulopathy, characterized by both hypercoagulable and hypocoagulable features. Similarly, ALI patients display a profound dysregulation of the fibrinolytic system with the vast majority of patients presenting with a hypofibrinolytic phenotype. Decades of research in experimental acute liver injury in mice suggest that fibrinolytic proteins, including plasmin(ogen), plasminogen activators, fibrinolysis inhibitors, and fibrin(ogen), can contribute to initial hepatotoxicity and/or stimulate liver repair. This review summarizes major experimental findings regarding the role of fibrinolytic factors in ALI from the last approximately 30 years and identifies unanswered questions, as well as highlighting areas for future research.
Collapse
Affiliation(s)
- Gina E Capece
- Department of Pharmacology, Rutgers University Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - Lauren G Poole
- Department of Pharmacology, Rutgers University Robert Wood Johnson Medical School, Piscataway, New Jersey
| |
Collapse
|
43
|
Shimizu J, Murao A, Lee Y, Aziz M, Wang P. Extracellular CIRP promotes Kupffer cell inflammatory polarization in sepsis. Front Immunol 2024; 15:1411930. [PMID: 38881891 PMCID: PMC11177612 DOI: 10.3389/fimmu.2024.1411930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/16/2024] [Indexed: 06/18/2024] Open
Abstract
Introduction Sepsis is a life-threatening inflammatory condition caused by dysregulated host responses to infection. Extracellular cold-inducible RNA-binding protein (eCIRP) is a recently discovered damage-associated molecular pattern that causes inflammation and organ injury in sepsis. Kupffer cells can be activated and polarized to the inflammatory M1 phenotype, contributing to tissue damage by producing proinflammatory mediators. We hypothesized that eCIRP promotes Kupffer cell M1 polarization in sepsis. Methods We stimulated Kupffer cells isolated from wild-type (WT) and TLR4-/- mice with recombinant mouse (rm) CIRP (i.e., eCIRP) and assessed supernatant IL-6 and TNFα levels by ELISA. The mRNA expression of iNOS and CD206 for M1 and M2 markers, respectively, was assessed by qPCR. We induced sepsis in WT and CIRP-/- mice by cecal ligation and puncture (CLP) and assessed iNOS and CD206 expression in Kupffer cells by flow cytometry. Results eCIRP dose- and time-dependently increased IL-6 and TNFα release from WT Kupffer cells. In TLR4-/- Kupffer cells, their increase after eCIRP stimulation was prevented. eCIRP significantly increased iNOS gene expression, while it did not alter CD206 expression in WT Kupffer cells. In TLR4-/- Kupffer cells, however, iNOS expression was significantly decreased compared with WT Kupffer cells after eCIRP stimulation. iNOS expression in Kupffer cells was significantly increased at 20 h after CLP in WT mice. In contrast, Kupffer cell iNOS expression in CIRP-/- mice was significantly decreased compared with WT mice after CLP. CD206 expression in Kupffer cells was not different across all groups. Kupffer cell M1/M2 ratio was significantly increased in WT septic mice, while it was significantly decreased in CIRP-/- mice compared to WT mice after CLP. Conclusion Our data have clearly shown that eCIRP induces Kupffer cell M1 polarization via TLR4 pathway in sepsis, resulting in overproduction of inflammatory cytokines. eCIRP could be a promising therapeutic target to attenuate inflammation by preventing Kupffer cell M1 polarization in sepsis.
Collapse
Affiliation(s)
- Junji Shimizu
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Yongchan Lee
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| |
Collapse
|
44
|
Young DJ, Edwards AJ, Quiroz Caceda KG, Liberzon E, Barrientos J, Hong S, Turner J, Choyke PL, Arlauckas S, Lazorchak AS, Morgan RA, Sato N, Dunbar CE. In vivo tracking of ex vivo generated 89 Zr-oxine labeled plasma cells by PET in a non-human primate model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595782. [PMID: 38903108 PMCID: PMC11188104 DOI: 10.1101/2024.05.24.595782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
B cells are an attractive platform for engineering to produce protein-based biologics absent in genetic disorders, and potentially for the treatment of metabolic diseases and cancer. As part of pre-clinical development of B cell medicines, we demonstrate a method to collect, ex vivo expand, differentiate, radioactively label, and track adoptively transferred non-human primate (NHP) B cells. These cells underwent 10- to 15-fold expansion, initiated IgG class switching, and differentiated into antibody secreting cells. Zirconium-89-oxine labeled cells were infused into autologous donors without any preconditioning and tracked by PET/CT imaging. Within 24 hours of infusion, 20% of the initial dose homed to the bone marrow and spleen and distributed stably and equally between the two. Interestingly, approximately half of the dose homed to the liver. Image analysis of the bone marrow demonstrated inhomogeneous distribution of the cells. The subjects experienced no clinically significant side effects or laboratory abnormalities. A second infusion of B cells into one of the subjects resulted in an almost identical distribution of cells, suggesting a non-limiting engraftment niche and feasibility of repeated infusions. This work supports the NHP as a valuable model to assess the potential of B cell medicines as potential treatment for human diseases.
Collapse
|
45
|
Cigalotto L, Martinvalet D. Granzymes in health and diseases: the good, the bad and the ugly. Front Immunol 2024; 15:1371743. [PMID: 38646541 PMCID: PMC11026543 DOI: 10.3389/fimmu.2024.1371743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Granzymes are a family of serine proteases, composed of five human members: GA, B, H, M and K. They were first discovered in the 1980s within cytotoxic granules released during NK cell- and T cell-mediated killing. Through their various proteolytic activities, granzymes can trigger different pathways within cells, all of which ultimately lead to the same result, cell death. Over the years, the initial consideration of granzymes as mere cytotoxic mediators has changed due to surprising findings demonstrating their expression in cells other than immune effectors as well as new intracellular and extracellular activities. Additional roles have been identified in the extracellular milieu, following granzyme escape from the immunological synapse or their release by specific cell types. Outside the cell, granzyme activities mediate extracellular matrix alteration via the degradation of matrix proteins or surface receptors. In certain contexts, these processes are essential for tissue homeostasis; in others, excessive matrix degradation and extensive cell death contribute to the onset of chronic diseases, inflammation, and autoimmunity. Here, we provide an overview of both the physiological and pathological roles of granzymes, highlighting their utility while also recognizing how their unregulated presence can trigger the development and/or worsening of diseases.
Collapse
Affiliation(s)
- Lavinia Cigalotto
- Laboratory of Reactive Oxygen Species and Cytotoxic Immunity, Department Biomedical Sciences, University of Padova, Padova, Italy
- Veneto Institute Of Molecular Medicine (VIMM), Padova, Italy
| | - Denis Martinvalet
- Laboratory of Reactive Oxygen Species and Cytotoxic Immunity, Department Biomedical Sciences, University of Padova, Padova, Italy
- Veneto Institute Of Molecular Medicine (VIMM), Padova, Italy
| |
Collapse
|
46
|
Zhao X, Yin F, Huang Y, Fu L, Ma Y, Ye L, Fan W, Gao W, Cai Y, Mou X. Oral administration of grape-derived nanovesicles for protection against LPS/D-GalN-induced acute liver failure. Int J Pharm 2024; 652:123812. [PMID: 38237707 DOI: 10.1016/j.ijpharm.2024.123812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 01/11/2024] [Accepted: 01/13/2024] [Indexed: 01/27/2024]
Abstract
Although the exploration of the molecular mechanisms of Acute liver failure (ALF) is supported by a growing number of studies, the lack of effective therapeutic agents and measures indicates an urgent clinical need for the development of new drugs and treatment strategies. Herein, we focused on the treatment of ALF with grape-derived nanovesicles (GDNVs), and assessed its protective effects and molecular mechanisms against liver injury. In the mice model of ALF, prophylactic administration for three consecutive days and treatment with GDNVs after successful induction of ALF showed a significant reduction of ALT and AST activity in mouse serum, as well as a blockade of the release of inflammatory cytokines IL6, IL-1β and TNF-α. Treatment with GDNVs significantly prevented the massive apoptosis of hepatocytes caused by LPS/D-GalN and down-regulated the activation and expression of the mitochondrial apoptosis-related proteins p53, Caspase 9, Caspase 8, Caspase 3 and Bax. GDNVs downregulated the release of chemokines during the inflammatory eruption in mice and inhibited the infiltration of peripheral monocytes into the liver by inhibiting CCR2/CCR5. Moreover, the pro-inflammatory phenotype of macrophages in the liver was reversed by GDNVs. GDNVs further reduced the activation of NLRP3-related pathways, and treatment with GDNVs activated the expression of autophagy-related proteins Foxo3a, Sirt1 and LC3-II in the damaged mouse liver, inducing autophagy to occur. GDNVs could exert hepatoprotective and inflammatory suppressive functions by increasing nuclear translocation of Nrf2 and upregulating HO-1 expression against exogenous toxin-induced oxidative stress in the liver. In conclusion, these results demonstrate that GDNVs alleviate LPS/D-GalN-induced ALF and have the potential to be used as a novel hepatoprotective agent for clinical treatment.
Collapse
Affiliation(s)
- Xin Zhao
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; College of Pharmacy, Hangzhou Medical College, Hangzhou 310059, China; Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Fang Yin
- Shanghai Engineering Research Center of Human Intestinal Microflora Function Development, Shanghai Tenth People's Hospital, Shanghai 200072, China
| | - Yilin Huang
- College of Pharmacy, Hangzhou Medical College, Hangzhou 310059, China
| | - Luoqin Fu
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Yingyu Ma
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Luyi Ye
- College of Pharmacy, Hangzhou Medical College, Hangzhou 310059, China
| | - Weijiao Fan
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Wenxue Gao
- Clinical Research Unit, Shanghai Tenth People's Hospital, Shanghai 200072, China.
| | - Yu Cai
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; College of Pharmacy, Hangzhou Medical College, Hangzhou 310059, China; Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China.
| | - Xiaozhou Mou
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; College of Pharmacy, Hangzhou Medical College, Hangzhou 310059, China; Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China.
| |
Collapse
|
47
|
Zhang Y, Wu D, Tian X, Chen B. From hepatitis B virus infection to acute-on-chronic liver failure: The dynamic role of hepatic macrophages. Scand J Immunol 2024; 99:e13349. [PMID: 38441398 DOI: 10.1111/sji.13349] [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: 08/26/2023] [Revised: 11/15/2023] [Accepted: 12/11/2023] [Indexed: 03/07/2024]
Abstract
Acute-on-chronic liver failure (ACLF) is a progressive disease that is associated with rapid worsening of clinical symptoms and high mortality. A multicentre prospective study from China demonstrated that patients with hepatitis B virus-related ACLF (HBV-ACLF) exhibited worse clinical characteristics and higher mortality rates compared to non-HBV-ACLF patients. Immune dysregulation is closely linked to the potential mechanisms of initiation and progression of ACLF. Innate immune response, which is represented by monocytes/macrophages, is up-regulated across ACLF development. This suggests that monocytes/macrophages play an essential role in maintaining the immune homeostasis of ACLF. Information that has been published in recent years shows that the immune status and function of monocytes/macrophages vary in ACLF precipitated by different chronic liver diseases. Monocytes/macrophages have an immune activation effect in hepatitis B-precipitated-ACLF, but they exhibit an immune suppression in cirrhosis-precipitated-ACLF. Therefore, this review aims to explain whether this difference affects the clinical outcome in HBV-ACLF patients as well as the mechanisms involved. We summarize the novel findings that highlight the dynamic polarization phenotype and functional status of hepatic macrophages from the stage of HBV infection to ACLF development. Moreover, we discuss how different HBV-related liver disease tissue microenvironments affect the phenotype and function of hepatic macrophages. In summary, increasing developments in understanding the differences in immune phenotype and functional status of hepatic macrophages in ACLF patients will provide new perspectives towards the effective restoration of ACLF immune homeostasis.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Hepatology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Dongsheng Wu
- Department of Anorectal Surgical, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Xiaoling Tian
- Department of Hepatology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Bin Chen
- Department of Hepatology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| |
Collapse
|
48
|
Eeda V, Patil NY, Joshi AD, Awasthi V. Advancements in metabolic-associated steatotic liver disease research: Diagnostics, small molecule developments, and future directions. Hepatol Res 2024; 54:222-234. [PMID: 38149861 PMCID: PMC10923026 DOI: 10.1111/hepr.14008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 12/28/2023]
Abstract
Metabolic (dysfunction)-associated steatotic liver disease (MASLD), formerly known as nonalcoholic fatty liver disease, is a growing global health concern with no approved pharmacological treatments. At the same time, there are no standard methods to definitively screen for the presence of MASLD because of its progressive nature and symptomatic commonality with other disorders. Recent advances in molecular understanding of MASLD pathophysiology have intensified research on development of new drug molecules, repurposing of existing drugs approved for other indications, and an educated use of dietary supplements for its treatment and prophylaxis. This review focused on depicting the latest advancements in MASLD research related to small molecule development for prophylaxis or treatment and diagnosis, with emphasis on mechanistic basis at the molecular level.
Collapse
Affiliation(s)
- Venkateswararao Eeda
- Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA
| | - Nikhil Yuvaraj Patil
- Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA
| | - Aditya Dilip Joshi
- Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA
| | - Vibhudutta Awasthi
- Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
49
|
Grandini NA, Costa MR, Gregolin CS, Siqueira JS, Vieira TA, Togneri Ferron AJ, Francisqueti-Ferron FV, Romualdo GR, Lúcia Dos Anjos Ferreira A, Aldini G, Corrêa CR, Moreto F. Effects of carnosine supplementation on markers for the pathophysiological development of metabolic dysfunction-associated steatotic liver disease in a diet-induced model. Mol Cell Endocrinol 2024; 582:112138. [PMID: 38147954 DOI: 10.1016/j.mce.2023.112138] [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: 09/21/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 12/28/2023]
Abstract
Consumption of diets high in sugar and fat is related to the development of Metabolic dysfunction-associated steatotic liver disease (MASLD). Carnosine (CAR) is a dipeptide with antioxidant and anti-inflammatory action and has been studied for treating diseases. This work aimed to evaluate the effects of CAR on diet-induced MASLD in rats. Male Wistar rats were distributed into 2 groups (17 weeks): normocaloric (Co, n = 12), and hypercaloric diet rich in lipids and simple carbohydrates (MASLD, n = 12). After, the animals were redistributed to begin the treatment with CAR (4 weeks): Co (n = 6), Co + CAR (n = 6), MASLD (n = 6), and MASLD + CAR (n = 6), administered intraperitoneally (250 mg/kg). Evaluations included nutritional, hormonal and metabolic parameters; hepatic steatosis, inflammatory and oxidative markers. MASLD group had a higher adiposity index, systolic blood pressure, glucose, plasma and liver triglycerides and cholesterol, insulin, hepatic steatosis, oxidative markers, and lower PPAR-α (Peroxisome Proliferator-activated receptor α), compared to the Co. CAR attenuated plasma and hepatic triglyceride and cholesterol levels, hepatic steatosis, CD68+ macrophages, and hepatic oxidative markers, in addition to increasing HDL cholesterol levels and PPAR-α, compared to the untreated MASLD group. CAR acts in importants pathophysiological processes of MASLD and may be a therapeutic compound to control the disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | | | - Fernando Moreto
- São Paulo State University (UNESP), Medical School, 18618687, Botucatu, Brazil
| |
Collapse
|
50
|
Lodge M, Scheidemantle G, Adams VR, Cottam MA, Richard D, Breuer D, Thompson P, Shrestha K, Liu X, Kennedy A. Fructose regulates the pentose phosphate pathway and induces an inflammatory and resolution phenotype in Kupffer cells. Sci Rep 2024; 14:4020. [PMID: 38369593 PMCID: PMC10874942 DOI: 10.1038/s41598-024-54272-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 02/10/2024] [Indexed: 02/20/2024] Open
Abstract
Over-consumption of fructose in adults and children has been linked to increased risk of non-alcoholic fatty liver disease (NAFLD). Recent studies have highlighted the effect of fructose on liver inflammation, fibrosis, and immune cell activation. However, little work summarizes the direct impact of fructose on macrophage infiltration, phenotype, and function within the liver. We demonstrate that chronic fructose diet decreased Kupffer cell populations while increasing transitioning monocytes. In addition, fructose increased fibrotic gene expression of collagen 1 alpha 1 (Col1a1) and tissue metallopeptidase inhibitor 1 (Timp1) as well as inflammatory gene expression of tumor necrosis factor alpha (Tnfa) and expression of transmembrane glycoprotein NMB (Gpnmb) in liver tissue compared to glucose and control diets. Single cell RNA sequencing (scRNAseq) revealed fructose elevated expression of matrix metallopeptidase 12 (Mmp12), interleukin 1 receptor antagonist (Il1rn), and radical S-adenosyl methionine domain (Rsad2) in liver and hepatic macrophages. In vitro studies using IMKC and J774.1 cells demonstrated decreased viability when exposed to fructose. Additionally, fructose increased Gpnmb, Tnfa, Mmp12, Il1rn, and Rsad2 in unpolarized IMKC. By mass spectrometry, C13 fructose tracing detected fructose metabolites in glycolysis and the pentose phosphate pathway (PPP). Inhibition of the PPP further increased fructose induced Il6, Gpnmb, Mmp12, Il1rn, and Rsad2 in nonpolarized IMKC. Taken together, fructose decreases cell viability while upregulating resolution and anti-inflammatory associated genes in Kupffer cells.
Collapse
Affiliation(s)
- Mareca Lodge
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, USA
| | - Grace Scheidemantle
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, USA
| | - Victoria R Adams
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, USA
| | - Matthew A Cottam
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Daniel Richard
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, USA
| | - Denitra Breuer
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, USA
| | - Peter Thompson
- Molecular Education, Technology and Research Innovation Center (METRIC), NC State University, Raleigh, NC, USA
| | - Kritika Shrestha
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, USA
| | - Xiaojing Liu
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, USA
| | - Arion Kennedy
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, USA.
| |
Collapse
|