1
|
Park N, Kim KS, Lee S, Choi JH, Na K. Enhanced stem cell-mediated therapeutic immune modulation with zinc oxide nanoparticles in liver regenerative therapy. Biomaterials 2025; 320:123232. [PMID: 40056611 DOI: 10.1016/j.biomaterials.2025.123232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/05/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
Liver regenerative therapy is critical for severe liver damage, including acute liver failure, fibrosis, post-cancer resection recovery, and autoimmune liver diseases, where restoration of liver tissues is essential. Stem cell-based therapies hold significant promise in liver regeneration by modulating immune responses to create a favorable healing microenvironment. However, their clinical efficacy has been limited by challenges such as poor cell engraftment and survival within the hostile injury site. To address these limitations, we developed a zinc oxide-derived nanoparticle (PZnONP) that enhances stem cell proliferation and activation by releasing bioactive Zn2+ and reactive oxygen species (ROS). Functionalized PZnONP exhibits pH-responsive behavior and improved dispersibility, enabling a lysosome-specific and sustained release of Zn2+ and ROS. Stem cells labeled with PZnONP (ZnBA) demonstrated anti-inflammatory properties, with paracrine effects influencing macrophages and damaged hepatocytes. In murine models of acute and fibrotic liver injury, it effectively migrated to the liver through stem cell homing effects and promoted anti-inflammatory responses by modulating Treg and Th17 cell polarization, as well as M2 and M1 macrophage balance, while reducing collagen synthesis. This study underscores the potential of integrating stem cell-based therapy with nanomedicine to improve regenerative outcomes in liver disease treatment.
Collapse
Affiliation(s)
- Naeun Park
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea; Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Kyoung Sub Kim
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea; Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Sanghee Lee
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jang Ho Choi
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea; Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Kun Na
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea; Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea.
| |
Collapse
|
2
|
Le PM, Pal-Ghosh S, Stepp MA, Menko AS. Shared Phenotypes of Immune Cells Recruited to the Cornea and the Surface of the Lens in Response to Formation of Corneal Erosions. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:960-981. [PMID: 39889825 PMCID: PMC12016862 DOI: 10.1016/j.ajpath.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/02/2025] [Accepted: 01/15/2025] [Indexed: 02/03/2025]
Abstract
Injuries to the cornea can lead to recurrent corneal erosions, compromising its barrier function and increasing the risk of infection. Vital as corneal integrity is to the eye's optical power and homeostasis, the immune response to corneal erosions remains poorly understood. It is also unknown whether there is coordinated immune activation between the cornea and other regions of the anterior segment to protect against microbial invasion and limit the spread of inflammation when corneal erosions occur. Herein, a corneal debridement wounding model was used to characterize the immune cell phenotypes populating the cornea in response to erosion formation, and whether and which immune cells are concurrently recruited to the surface of the lens was investigated. The formation of corneal erosions induced an influx of myeloid lineage phenotypes, both M2 macrophages associated with tissue healing and wound repair, and Ly6G+ Ly6C+ myeloperoxidase+ cells resembling neutrophils/polymorphonuclear-myeloid-derived suppressor cells (PMN-MDSCs), with few regulatory T cells, into the corneal stroma under erosion sites. This leukocyte migration into the cornea when erosions develop was paralleled by the recruitment of immune cells, predominantly neutrophils/PMN-MDSCs, to the anterior, cornea-facing lens capsule. Both cornea-infiltrating and lens capsule-associated neutrophil/PMN-MDSC-like immune cells produce the anti-inflammatory cytokine IL-10. These findings suggest a collaborative role for the lens capsule-associated immune cells in preventing infections, controlling inflammation, and maintaining homeostasis of the anterior segment during recurrent corneal erosions.
Collapse
Affiliation(s)
- Phuong M Le
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sonali Pal-Ghosh
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Mary Ann Stepp
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia; Department of Ophthalmology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - A Sue Menko
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania; Department of Ophthalmology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
3
|
Ghosh C, Kundu T, Pathak T, Saini S, Das N, Saini S, Sircar D, Kumar P, Roy P. Indian lychee honey ameliorates hepatic glucose uptake by regulating the ChREBP/Glut4 axis under insulin-resistant conditions. Food Funct 2025; 16:2031-2056. [PMID: 39963045 DOI: 10.1039/d4fo03900a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Many traditional treatments include honey owing to its magnificent health beneficiary effects. Recent studies have demonstrated the potent anti-diabetic activity of honey. However, its actual mechanism of action remains elusive. Moreover, being rich in sugar (75%-80%), its role in maintaining glucose homeostasis remains questionable. Although the polyphenol content of honey aids its hypoglycaemic activity, the small quantity of bioactive compounds in honey (0.5%-1.0%) may not be solely responsible for this. In the current study, an attempt was made to understand the role of Indian lychee honey (LyH) in regulating blood glucose levels under diabetic conditions. This study investigated whether LyH, although rich in sugars, can be used as an alternative to regulate glucose and lipid homeostasis under insulin-resistant conditions by regulating the ChREBP/Glut4 signalling pathway. This study was first performed in vitro in palmitic acid-induced insulin-resistant HepG2 cells. Various assays, such as FACS, GCMS, qRT-PCR, immunoblot and ChIP-qPCR, were performed to establish the anti-hyperglycaemic role of LyH in vitro. The in vitro results were subsequently confirmed in vivo using a high-fat diet-induced diabetic C57BL/6 mice model. The in vivo study was supported by several experiments, such as examining blood parameters, histopathology, double-immunohistochemistry and ELISA. Finally, the finding was validated by comparing it with a couple of GEO datasets from the NCBI database. This study found that LyH is an excellent choice for regulating blood sugar levels under diabetic conditions without significant harmful side effects. Moreover, LyH showed excellent hepatic glucose uptake activity in an insulin-independent manner. This activity is mainly governed by sugars as its main ingredient. LyH treatment also regulates hepatic lipid homeostasis by maintaining a balance between saturated and unsaturated fatty acids in insulin-resistant HepG2 cells. Further, sugar, when supplemented individually, caused severe inflammation, which was validated through histopathology, ELISA and IHC. Collectively, the findings of this study indicate that Indian LyH provides a better food matrix (the right proportion of sugars and different bioactive compounds), which significantly improves hyperglycemia and inflammation under diabetic conditions by regulating the hepatic ChREBP/Glut4 axis.
Collapse
Affiliation(s)
- Chandrachur Ghosh
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
| | - Tathagata Kundu
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
| | - Tiyasa Pathak
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
| | - Saakshi Saini
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
| | - Neeladrisingha Das
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
| | - Surendra Saini
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
| | - Debabrata Sircar
- Plant Molecular Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India
| | - Prabhat Kumar
- National Bee Board, DA & FW, Ministry of Agriculture and Farmers Welfare, B Wing, 2nd Floor, Janpath Bhawan, Janpath, New Delhi - 110 001, India
| | - Partha Roy
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
- Center for Indian Knowledge Systems, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India
| |
Collapse
|
4
|
Entsie P, Amoafo EB, Kang Y, Gustad T, Dorsam GP, Frey MR, Liverani E. Sex-specific activation of platelet purinergic signaling is key in local cytokine release and phagocytosis in the peritoneal cavity in intra-abdominal sepsis. Am J Physiol Cell Physiol 2025; 328:C791-C805. [PMID: 39854048 DOI: 10.1152/ajpcell.00116.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/07/2024] [Accepted: 01/17/2025] [Indexed: 01/26/2025]
Abstract
Intra-abdominal sepsis is a life-threatening complex syndrome caused by microbes in the gut microbiota invading the peritoneal cavity. It is one of the major complications of intra-abdominal surgery. To date, only supportive therapies are available. No studies have investigated the progression of intra-abdominal sepsis in the peritoneal cavity. Our group has shown that platelets play an essential role during sepsis, and blocking purinergic signaling in platelets through P2Y1 and P2Y12 antagonism significantly lowered inflammatory levels and improved survival in a murine model of sepsis. Here, we tested whether antagonizing purinergic signaling in platelets in the peritoneal cavity can reduce the local release of cytokines and modulate platelet interaction with the immune system. We used cecal ligation and puncture (CLP) to induce sepsis followed by intraperitoneal administration of MRS2279 (P2Y1 antagonist) or ticagrelor (P2Y12 antagonist) in male and female mice. The peritoneal cavity fluid (PCF) was collected 4 or 24 h post-CLP and analyzed for cell recruitment, platelet markers, cytokines, and platelet immune cell interactions. Platelet markers were increased 24 h after CLP, although the total platelet count in the peritoneal cavity was lower than the blood. Blocking P2Y12 or P2Y1 improved bacterial clearance in the PCF in a sex-dependent manner. The influx of immune cells in the peritoneal cavity was altered by blocking P2Y12 or P2Y1 sex-dependently. Blocking P2Y1 and P2Y12 receptors can enhance the phagocytic activity in the peritoneal cavity in a sex- and time-related manner, and platelets significantly contribute to the development and progression of sepsis in the peritoneal cavity.NEW & NOTEWORTHY Intra-abdominal sepsis is a challenging complication postabdominal surgery caused by perforations of the gastrointestinal tract where microbes invade the peritoneal cavity. This leads to local cytokine release and immune cell dysfunction. Our data identify platelets as key players in mediating inflammation in intra-abdominal sepsis. We have shown that blocking purinergic signaling in the peritoneal cavity reduced cytokine release and cell-cell interactions differently in males and females, hence a sex-specific strategy to improve intra-abdominal sepsis.
Collapse
Affiliation(s)
- Philomena Entsie
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Emmanuel Boadi Amoafo
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Ying Kang
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Thomas Gustad
- Department of Microbiological Sciences, College of Agriculture, Food Systems and Natural Resources, North Dakota State University, Fargo, North Dakota, United States
| | - Glenn P Dorsam
- Department of Microbiological Sciences, College of Agriculture, Food Systems and Natural Resources, North Dakota State University, Fargo, North Dakota, United States
| | - Mark R Frey
- Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| | - Elisabetta Liverani
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| |
Collapse
|
5
|
Kuklin A, Slabber CF, Tortola L, Kwan CL, Liebisch G, Kondylis V, Mair F, Kopf M, Weber A, Werner S. An Nrf2-NF-κB Crosstalk Controls Hepatocyte Proliferation in the Normal and Injured Liver. Cell Mol Gastroenterol Hepatol 2025; 19:101480. [PMID: 39970988 DOI: 10.1016/j.jcmgh.2025.101480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND & AIMS The liver has remarkable regenerative and detoxification capacities, which require the Nrf2 and NF-κB transcription factors. Although their individual functions in hepatocytes are well characterized, knowledge about their crosstalk in the adult liver is limited. METHODS We performed AAV8-Cre inducible, hepatocyte-specific knockout of Nrf2, the NF-κB subunit p65, or both genes to determine the individual and combined roles of these transcription factors in the intact liver of male adult mice and after acute CCl4 injury. Mice were characterized using histologic and immunohistochemical stainings, serum and liver bile acid analysis, flow cytometry, and RNA sequencing. To distinguish between cell-autonomous and non-cell-autonomous mechanisms, we generated and analyzed knockout and knockdown AML12 liver cells. Clodronate liposome-mediated macrophage depletion was used to determine the role of these immune cells in hepatocyte proliferation after CCl4 injection. RESULTS Loss of p65 alone or p65 in combination with Nrf2 caused spontaneous liver inflammation and necrosis. Gene expression profiling identified individual and common target genes of both transcription factors, including genes involved in the control of cell proliferation. Consistent with the expression of these genes, hepatocyte proliferation was reduced by Nrf2 deficiency under homeostatic conditions and after CCl4 injury, which was rescued by additional loss of p65. The increased hepatocyte proliferation in the double-knockout mice was non-cell-autonomous and correlated with macrophage accumulation in the liver. Depletion of macrophages in these mice suppressed hepatocyte proliferation after CCl4 treatment. CONCLUSIONS These results reveal a crosstalk between Nrf2 and p65 in the control of hepatocyte proliferation and point to a key role of macrophages in this effect.
Collapse
Affiliation(s)
- Andrii Kuklin
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland.
| | | | - Luigi Tortola
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Chan Lap Kwan
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital Zurich, Zurich, Switzerland; Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Vangelis Kondylis
- Department of Gastroenterology, Hepatology and Infectious Diseases, University of Düsseldorf, Düsseldorf, Germany
| | - Florian Mair
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital Zurich, Zurich, Switzerland; Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
6
|
Zhang W, Zeng H, Xie S, Yu C, Zhang M, Chen Q, Dong H, Zhang H, Lin H, Zheng N, Zhu L, Lu J. Activation of autophagy with PF-06409577 alleviates heatstroke-induced organ injury. ENVIRONMENT INTERNATIONAL 2025; 196:109285. [PMID: 39855028 DOI: 10.1016/j.envint.2025.109285] [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: 10/03/2024] [Revised: 12/23/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
Heat waves are a significant environmental issue threatening global human health. Extreme temperatures can lead to various heat-related illnesses, with heatstroke being among the most severe. Currently, there are no effective treatments to mitigate the multi-organ damage caused by heatstroke. We found that heat stress activated autophagy. Knockdown of the autophagy-related gene 7 (ATG7) or knockout of the autophagy initiation regulatory genes UNC-51-like autophagy activating kinase 1/2 (ULK1/ULK2) increased cell death. PF-06409577, an allosteric activator of AMP-activated protein kinase β (AMPKβ), reduced heat stress-induced cell death by promoting autophagy. Inhibition of ATG7 or ULK1 weakened PF-06409577's protective effect on cells. Treatment of heatstroke mouse models with PF-06409577 suppressed high temperature-induced damage to multiple organs, including the liver, kidneys, lungs, and small intestine. PF-06409577 protected liver and kidney functions, lowered the expression of kidney injury markers neutrophil gelatinase associated lipocalin (Ngal), secreted phosphoprotein 1 (Spp1), and clusterin (Clu), and reduced levels of the inflammatory factor IL-6. Additionally, it decreased heat stress-induced macrophage infiltration and IL-6 production in the liver. The results indicate that activation of autophagy serves a protective function during heat stress, and the AMPK activator PF-06409577 exhibits potential in mitigating heatstroke-induced multi-organ damage through its ability to promote autophagy.
Collapse
Affiliation(s)
- Wei Zhang
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzong Teaching Hospital (900th Hospital of Joint Logistic Support Force), Fujian University of Traditional Chinese Medicine, Fuzhou 350025, China
| | - Huajing Zeng
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China
| | - Siyu Xie
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzong Teaching Hospital (900th Hospital of Joint Logistic Support Force), Fujian University of Traditional Chinese Medicine, Fuzhou 350025, China; Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China; Dongfang Hospital, Xiamen University, Fuzhou 350025, China
| | - Cheng Yu
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzong Teaching Hospital (900th Hospital of Joint Logistic Support Force), Fujian University of Traditional Chinese Medicine, Fuzhou 350025, China; Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China; Dongfang Hospital, Xiamen University, Fuzhou 350025, China
| | - Meina Zhang
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzong Teaching Hospital (900th Hospital of Joint Logistic Support Force), Fujian University of Traditional Chinese Medicine, Fuzhou 350025, China; Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China; Dongfang Hospital, Xiamen University, Fuzhou 350025, China
| | - Qiuyan Chen
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzong Teaching Hospital (900th Hospital of Joint Logistic Support Force), Fujian University of Traditional Chinese Medicine, Fuzhou 350025, China; Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China; Dongfang Hospital, Xiamen University, Fuzhou 350025, China
| | - Huiyue Dong
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzong Teaching Hospital (900th Hospital of Joint Logistic Support Force), Fujian University of Traditional Chinese Medicine, Fuzhou 350025, China; Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China; Dongfang Hospital, Xiamen University, Fuzhou 350025, China
| | - Hui Zhang
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzong Teaching Hospital (900th Hospital of Joint Logistic Support Force), Fujian University of Traditional Chinese Medicine, Fuzhou 350025, China; Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China; Dongfang Hospital, Xiamen University, Fuzhou 350025, China
| | - Hao Lin
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China
| | - Nengjing Zheng
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzong Teaching Hospital (900th Hospital of Joint Logistic Support Force), Fujian University of Traditional Chinese Medicine, Fuzhou 350025, China
| | - Lin Zhu
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzong Teaching Hospital (900th Hospital of Joint Logistic Support Force), Fujian University of Traditional Chinese Medicine, Fuzhou 350025, China; Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China; Dongfang Hospital, Xiamen University, Fuzhou 350025, China
| | - Jun Lu
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzong Teaching Hospital (900th Hospital of Joint Logistic Support Force), Fujian University of Traditional Chinese Medicine, Fuzhou 350025, China; Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China; Dongfang Hospital, Xiamen University, Fuzhou 350025, China; Organ Transplant Institute, 900th Hospital of Joint Logistic Support Force, Fuzhou 350025, China.
| |
Collapse
|
7
|
Humphries C, Addison ML, Dear JW, Forbes SJ. The emerging role of alternatively activated macrophages to treat acute liver injury. Arch Toxicol 2025; 99:103-114. [PMID: 39503878 PMCID: PMC11742291 DOI: 10.1007/s00204-024-03892-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/09/2024] [Indexed: 01/19/2025]
Abstract
Acute liver injury (ALI) has a clear requirement for novel therapies. One emerging option is the use of alternatively activated macrophages (AAMs); a distinct subtype of macrophage with a role in liver injury control and repair. In this comprehensive review, we provide an overview of the current limited options for ALI, and the potential advantages offered by AAMs. We describe the evidence supporting their use from in vitro studies, pre-clinical animal studies, and human clinical trials. We suggest why the first evidence for the clinical use of AAMs is likely to be found in acetaminophen toxicity, and discuss the specific evidence for AAM use in this population, as well as potential applications for AAMs in other patient populations. The key domains by which the performance of AAMs for the treatment of ALI will be assessed are identified, and remaining challenges to the successful delivery of AAMs to clinic are explored.
Collapse
Affiliation(s)
- Chris Humphries
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Drive, Edinburgh, UK
| | - Melisande L Addison
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Drive, Edinburgh, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - James W Dear
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Drive, Edinburgh, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
8
|
Sun Y, He X, Han J, Yin W, Wang H, Li J, Liu W, Kuai X, Lv J, Ji J. Activated hepatic stellate cell-derived small extracellular vesicles facilitate M2 macrophage polarization and hepatoma progression via miR-27a-3p. Front Immunol 2024; 15:1489679. [PMID: 39742261 PMCID: PMC11685157 DOI: 10.3389/fimmu.2024.1489679] [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: 09/01/2024] [Accepted: 12/03/2024] [Indexed: 01/03/2025] Open
Abstract
The progression of hepatoma is heavily influenced by the microenvironment. Tumor-associated macrophages (TAMs) are considered to play a critical role in the tumor microenvironment (TME) and increase the aggressiveness of hepatoma. The activation of hepatic stellate cells (HSCs) is involved in hepatoma progression, and accumulating evidence demonstrates a change in microRNA (miRNA) expression during HSC activation. Therefore, the potential roles of HSCs-related miRNAs in macrophage differentiation and hepatoma progression deserve to be explored. The present study aimed to investigate the effects of miRNAs carried by small extracellular vesicles (sEVs) released by activated HSCs on hepatoma progression. The results indicated that miR-27a-3p was significantly upregulated in cells and corresponding sEVs during the activation of primary rat HSCs and human HSC line-LX2 cells. Furthermore, miR-27a-3p contributed to the proliferation and migration of hepatoma cells and promoted M2 polarization of macrophage. HSC-sEVs overexpressing miR-27a-3p can directly facilitate tumor progression and modulate macrophage polarization, indirectly contributing to hepatoma progression. Finally, Sprouty2 (SPRY2) was verified to be the target gene of miR-27a-3p. In conclusion, activated HSC-derived sEVs with high levels of miR-27a-3p might induce M2 macrophage polarization and promote hepatoma progression, providing new insights into the mechanism of hepatoma progression.
Collapse
Affiliation(s)
- Yufeng Sun
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| | - Xiaoqian He
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| | - Jiayi Han
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| | - Wenxuan Yin
- Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Haichen Wang
- Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Jing Li
- Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Weiqi Liu
- Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Xingwang Kuai
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| | - Jiaying Lv
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| | - Juling Ji
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, China
| |
Collapse
|
9
|
Macirella R, Ahmed AIM, Talarico F, Gharbi N, Mezzasalma M, Brunelli E. Morphological Alterations and Oxidative Stress Induction in Danio rerio Liver After Short-Term Exposure to the Strobilurin Fungicide Dimoxystrobin. ENVIRONMENTS 2024; 11:282. [DOI: 10.3390/environments11120282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Unlike many other fungicides, strobilurins are applied several times during the growing season for prophylactic purposes, thus heightening the risk of environmental contamination. In the EU, the dimoxystrobin approval period lasted for 17 years. It has been classified as moderately toxic to birds and highly toxic to earthworms, and it is suspected to be carcinogenic to humans. However, it is still commercialized in several countries. The effects of dimoxystrobin are still largely underexplored, with only three studies reporting sublethal alterations in fish. Here, we evaluated for the first time the effects of dimoxystrobin on zebrafish liver after short-term exposure (96 h) to two sublethal and environmentally relevant concentrations (6.56 and 13.13 μg/L), providing evidence of morphological, functional, and ultrastructural modifications. We revealed severe alterations encompassing three reaction patterns: circulatory disturbance, regressive and progressive changes, which also showed a dose-dependent trend. Furthermore, we revealed that dimoxystrobin induced a significant increase in lipid content, a decrease in glycogen granules and affected the defensive response against oxidative stress through a significant downregulation of SOD and CAT. The information presented here demonstrates that the hazardous properties of dimoxystrobin may result from several pathological events involving multiple targets. Our results also emphasize the importance of the combined use of morphological, ultrastructural and functional investigation in ecotoxicological studies.
Collapse
Affiliation(s)
- Rachele Macirella
- Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, Via P. Bucci 4/B, 87036 Rende, Italy
| | - Abdalmoiz I. M. Ahmed
- Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, Via P. Bucci 4/B, 87036 Rende, Italy
| | - Federica Talarico
- Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, Via P. Bucci 4/B, 87036 Rende, Italy
- Natural History Museum and Botanical Garden, University of Calabria, 87036 Rende, Italy
| | - Naouel Gharbi
- Fish Biology and Aquaculture Group, Ocean and Environment Department, NORCE Norwegian Research Center, 5006 Bergen, Norway
| | - Marcello Mezzasalma
- Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, Via P. Bucci 4/B, 87036 Rende, Italy
| | - Elvira Brunelli
- Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, Via P. Bucci 4/B, 87036 Rende, Italy
| |
Collapse
|
10
|
Lam P, Zygmunt DA, Ashbrook A, Yan C, Du H, Martin PT. Liver-directed AAV gene therapy normalizes disease symptoms and provides cross-correction in a model of lysosomal acid lipase deficiency. Mol Ther 2024; 32:4272-4284. [PMID: 39489913 PMCID: PMC11638878 DOI: 10.1016/j.ymthe.2024.10.022] [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: 06/20/2024] [Revised: 09/11/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
Lysosomal acid lipase deficiency (LAL-D) is caused by mutations in the LIPA gene, which encodes the lysosomal enzyme that hydrolyzes triglycerides and cholesteryl esters to free fatty acids and free cholesterol. The objective of this study was to develop a curative single-treatment therapy for LAL-D using adeno-associated virus (AAV). Treatment at both early (1-2 days) and late (8-week) timepoints with rscAAVrh74.LP1.LIPA, a liver-directed AAV gene therapy, normalized many disease measures in Lipa-/- mice when measured at 24 weeks of age, including hepatosplenomegaly, serum transaminase activity, organ triglyceride and cholesterol levels, and biomarkers of liver inflammation and fibrosis. For most measures, liver-directed therapy was superior to therapy utilizing a constitutive tissue expression approach. rscAAVrh74.LP1.LIPA treatment elevated LAL enzyme activity above wild-type levels in all tissues tested, including liver, spleen, intestine, muscle, and brain, and treatment elicited minimal serum antibody responses to transgenic protein. AAV treatment at 8 weeks of age with 1 × 1013 vg/kg extended survival significantly, with all AAV-treated mice surviving beyond the maximal lifespan of untreated Lipa-/- mice. These results show that this liver-directed LIPA gene therapy has the potential to be a transformative treatment for LAL-D.
Collapse
Affiliation(s)
- Patricia Lam
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA
| | - Deborah A Zygmunt
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA
| | - Anna Ashbrook
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA
| | - Cong Yan
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hong Du
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Paul T Martin
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA; Department of Pediatrics, The Ohio State University School of Medicine, Columbus, OH 43210, USA.
| |
Collapse
|
11
|
Ma X, Qiu J, Zou S, Tan L, Miao T. The role of macrophages in liver fibrosis: composition, heterogeneity, and therapeutic strategies. Front Immunol 2024; 15:1494250. [PMID: 39635524 PMCID: PMC11616179 DOI: 10.3389/fimmu.2024.1494250] [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: 09/10/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Macrophages, the predominant immune cells in the liver, are essential for maintaining hepatic homeostasis and responding to liver injury caused by external stressors. The hepatic macrophage population is highly heterogeneous and plastic, mainly comprised of hepatic resident kuffer cells (KCs), monocyte-derived macrophages (MoMφs), lipid-associated macrophages (LAMs), and liver capsular macrophages (LCMs). KCs, a population of resident macrophages, are localized in the liver and can self-renew through in situ proliferation. However, MoMφs in the liver are recruited from the periphery circulation. LAMs are a self-renewing subgroup of liver macrophages near the bile duct. While LCMs are located in the liver capsule and derived from peripheral monocytes. LAMs and LCMs are also involved in liver damage induced by various factors. Hepatic macrophages exhibit distinct phenotypes and functions depending on the specific microenvironment in the liver. KCs are critical for initiating inflammatory responses after sensing tissue damage, while the MoMφs infiltrated in the liver are implicated in both the progression and resolution of chronic hepatic inflammation and fibrosis. The regulatory function of liver macrophages in hepatic fibrosis has attracted significant interest in current research. Numerous literatures have documented that the MoMφs in the liver have a dual impact on the progression and resolution of liver fibrosis. The MoMφs in the liver can be categorized into two subtypes based on their Ly-6C expression level: inflammatory macrophages with high Ly-6C expression (referred to as Ly-6Chi subgroup macrophages) and reparative macrophages with low Ly-6C expression (referred to as Ly-6Clo subgroup macrophages). Ly-6Chi subgroup macrophages are conducive to the occurrence and progression of liver fibrosis, while Ly-6Clo subgroup macrophages are associated with the degradation of extracellular matrix (ECM) and regression of liver fibrosis. Given this, liver macrophages play a pivotal role in the occurrence, progression, and regression of liver fibrosis. Based on these studies, treatment therapies targeting liver macrophages are also being studied gradually. This review aims to summarize researches on the composition and origin of liver macrophages, the macrophage heterogeneity in the progression and regression of liver fibrosis, and anti-fibrosis therapeutic strategies targeting macrophages in the liver.
Collapse
Affiliation(s)
- Xiaocao Ma
- Department of Nuclear Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jia Qiu
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Intelligent Medical Imaging of Jiangxi Key Laboratory, Nanchang, China
| | - Shubiao Zou
- Department of Nuclear Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Liling Tan
- Department of Nuclear Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Tingting Miao
- Department of Nuclear Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
12
|
Jung H, Kyun ML, Kwon JI, Kim J, Kim JK, Park D, Lee YB, Moon KS. Amplified response of drug-induced liver fibrosis via immune cell co-culture in a 3D in vitro hepatic fibrosis model. Biomater Sci 2024. [PMID: 39483068 DOI: 10.1039/d4bm00874j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Liver fibrosis, a critical consequence of chronic liver diseases, is characterized by excessive extracellular matrix (ECM) deposition driven by inflammation. This process involves complex interactions among hepatocytes, hepatic stellate cells (HSCs), and Kupffer cells, the liver's resident macrophages. Kupffer cells are essential in initiating fibrosis through the release of pro-inflammatory cytokines that activate HSCs. Although various in vitro liver fibrosis models have been developed, there is a lack of models that include the immune environment of the liver to clarify the influence of immune cells on the progression of liver fibrosis. We developed an in vitro liver fibrosis model by co-culturing hepatocytes (HepaRG), a hepatic stellate cell line (LX-2), and macrophages (differentiated THP-1). The effects of liver fibrosis inducers, transforming growth factor-beta1 (TGF-β1) and methotrexate (MTX), on the inflammatory response and stellate cell activation were evaluated in this triple co-culture model. A triple co-culture condition was developed as a 3D in vitro model using gelatin methacrylate (GelMA), offering a more biomimetic environment and achieving liver fibrosis via immune cell activation associated ECM deposition. In this study, the developed triple co-culture model has the potential to elucidate cell progression roles in liver fibrosis and can be applied in drug screening and toxicity assessments targeting liver fibrosis.
Collapse
Affiliation(s)
- Hyewon Jung
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
- Human and Environmental Toxicology, University of Science and Technology, Daejeon, 34114, Republic of Korea.
| | - Mi-Lang Kyun
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
| | - Ji-In Kwon
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
- Department of Food and Nutrition, University of Hannam, Daejeon, 34054, Republic of Korea
| | - Jeongha Kim
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
- Department of Food and Nutrition, University of Hannam, Daejeon, 34054, Republic of Korea
| | - Ju-Kang Kim
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
| | - Daeui Park
- Human and Environmental Toxicology, University of Science and Technology, Daejeon, 34114, Republic of Korea.
- Center for Biomimetic Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Yu Bin Lee
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
- Human and Environmental Toxicology, University of Science and Technology, Daejeon, 34114, Republic of Korea.
| | - Kyoung-Sik Moon
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
- Human and Environmental Toxicology, University of Science and Technology, Daejeon, 34114, Republic of Korea.
| |
Collapse
|
13
|
Liu G, Das SK. D-Xylose Ameliorates Non-Alcoholic Fatty Liver Disease by Targeting Macrophage-expressed LYZ Gene. Cell Biochem Biophys 2024:10.1007/s12013-024-01572-7. [PMID: 39379786 DOI: 10.1007/s12013-024-01572-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2024] [Indexed: 10/10/2024]
Abstract
This study investigates the therapeutic effects of D-Xylose, a natural sugar, on non-alcoholic fatty liver disease (NAFLD), focusing on the expression of the lysozyme gene (LYZ) in macrophages. Using the single-cell dataset GSE136103 for NAFLD, researchers analyzed macrophage populations and other groups utilizing the Seurat package in R, while a differential analysis was performed on the NAFLD dataset GSE61260 using the limma package. Both in vitro and in vivo models, including cell culture, mouse models, RT-qPCR, Western blot, ELISA, and histopathological analyses, were employed to examine the effect of D-Xylose on lipid accumulation, LYZ expression, blood lipid levels, and inflammatory responses. The study found a significant upregulation of LYZ in free fatty acid (FFA)-treated cells and mouse liver tissues, with a subsequent reduction after D-Xylose intervention. Treatment with D-Xylose and Amlodipine led to a notable decrease in lipid accumulation, as evidenced by reduced triglyceride and cholesterol levels. D-Xylose demonstrated a greater improvement in lipid metabolism than Amlodipine. Additionally, D-Xylose significantly mitigated inflammatory responses, reducing levels of inflammatory markers such as IL1R, IL6, MYS8, TNF, NF-κB, and IL-1. Furthermore, D-Xylose administration significantly reduced liver weight and liver index, with a positive impact on serum liver function and blood lipid levels. The findings suggest that D-Xylose could be a therapeutic intervention for NAFLD by targeting LYZ expression in macrophages, thereby modulating lipid metabolism and inflammatory responses.
Collapse
Affiliation(s)
- Guoxiang Liu
- Faculty of Pharmacy, Lincoln University College, Petaling Jaya, Selangor, Malaysia
| | - Sreemoy Kanti Das
- Faculty of Pharmacy, Lincoln University College, Petaling Jaya, Selangor, Malaysia.
| |
Collapse
|
14
|
Torabi S, Zarrabi M, Shekari F, Poorkazem H, Lotfinia M, Bencina S, Gramignoli R, Hassan M, Najimi M, Vosough M. Wharton's Jelly mesenchymal stem cell-derived extracellular vesicles induce liver fibrosis-resolving phenotype in alternatively activated macrophages. J Cell Mol Med 2024; 28:e18507. [PMID: 39288445 PMCID: PMC11407755 DOI: 10.1111/jcmm.18507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 09/19/2024] Open
Abstract
The potential of extracellular vesicles (EVs) isolated from mesenchymal stromal cells in guiding macrophages toward anti-inflammatory immunophenotypes, has been reported in several studies. In our study, we provided experimental evidence of a distinctive effect played by Wharton Jelly mesenchymal stromal cell-derived EVs (WJ-EVs) on human macrophages. We particularly analyzed their anti-inflammatory effects on macrophages by evaluating their interactions with stellate cells, and their protective role in liver fibrosis. A three-step gradient method was used to isolate monocytes from umbilical cord blood (UCB). Two subpopulations of WJ-EVs were isolated by high-speed (20,000 g) and differential ultracentrifugation (110,000 g). Further to their characterization, they were designated as EV20K and EV110K and incubated at different concentrations with UCB-derived monocytes for 7 days. Their anti-fibrotic effect was assessed by studying the differentiation and functional levels of generated macrophages and their potential to modulate the survival and activity of LX2 stellate cells. The EV20K triggers the polarization of UCB-derived monocytes towards a peculiar M2-like functional phenotype more effectively than the M-CSF positive control. The EV20K treated macrophages were characterized by a higher expression of scavenger receptors, increased phagocytic capacity and production level of interleukin-10 and transforming growth factor-β. Conditioned medium from those polarized macrophages attenuated the proliferation, contractility and activation of LX2 stellate cells. Our data show that EV20K derived from WJ-MSCs induces activated macrophages to suppress immune responses and potentially play a protective role in the pathogenesis of liver fibrosis by directly inhibiting HSC's activation.
Collapse
Affiliation(s)
- Shukoofeh Torabi
- Department of Regenerative Medicine, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in MedicineMazandaran University of Medical SciencesSariIran
| | - Morteza Zarrabi
- Department of Regenerative Medicine, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Hedie Poorkazem
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Majid Lotfinia
- Physiology Research CenterKashan University of Medical SciencesKashanIran
| | - Stefan Bencina
- Department of Laboratory Medicine, Division of PathologyKarolinska InstitutetStockholmSweden
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of PathologyKarolinska InstitutetStockholmSweden
- UOSD Cell FactoryIRCCS Istituto Giannina GasliniGenoaItaly
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine and Karolinska University HospitalKarolinska InstituteStockholmSweden
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell TherapyInstitute of Experimental and Clinical Research (IREC), UCLouvainBrusselsBelgium
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Experimental Cancer Medicine, Institution for Laboratory Medicine and Karolinska University HospitalKarolinska InstituteStockholmSweden
| |
Collapse
|
15
|
Brenøe JE, van Hoorn EGM, Beck L, Bulthuis M, Bezemer RE, Gordijn SJ, Schoots MH, Prins JR. Altered placental macrophage numbers and subsets in pregnancies complicated with intrahepatic cholestasis of pregnancy (ICP) compared to healthy pregnancies. Placenta 2024; 153:22-30. [PMID: 38810541 DOI: 10.1016/j.placenta.2024.05.129] [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: 10/30/2023] [Revised: 04/29/2024] [Accepted: 05/15/2024] [Indexed: 05/31/2024]
Abstract
INTRODUCTION Intrahepatic cholestasis of pregnancy (ICP) can result in adverse outcomes for both mother and fetus. Inflammatory (M1 subset) or anti-inflammatory (M2 subset) macrophage polarisation is associated with various complications of pregnancy. However, the influence of ICP on macrophage numbers and polarisation remains unknown. This study analyses macrophage density and distribution in placentas of patients with ICP compared to controls. Clinical parameters were correlated to macrophage distribution and ursodeoxycholic acid use (UDCA). METHODS This study included routinely collected placental tissue samples of 42 women diagnosed with ICP and of 50 control pregnancies. Immunohistochemical staining was performed on placental tissue using CD68 antibody as a pan-macrophage marker, CD206 antibody as an M2 and HLA-DR antibody as an M1 macrophage marker. Macrophage density (cells/mm2) and distribution (CD206+/CD68+ or CD206+/CD68+HLA-DR+) in both decidua (maternal tissue) and villous parenchyma (fetal tissue) were compared between groups. Macrophage density and distribution were correlated to clinical parameters for ICP patients. RESULTS The density of CD68+ macrophages differed significantly between groups in villous parenchyma. In both decidua and villous parenchyma, CD206+/CD68+ ratio was significantly lower in ICP patients compared to controls (p = 0.003 and p=<0.001, respectively). No difference was found based on UDCA use or in CD68+HLA-DR+ cell density. Significant correlations were found between macrophage density and peak serum bile acids and liver enzymes. DISCUSSION In ICP patients, an immune shift was observed in both decidual and villous tissue, indicated by a lower CD206+/CD68+ ratio. ICP seems to affect placental tissue, however more research is required to understand its consequences.
Collapse
Affiliation(s)
- J E Brenøe
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - E G M van Hoorn
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - L Beck
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - M Bulthuis
- Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - R E Bezemer
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - S J Gordijn
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - M H Schoots
- Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - J R Prins
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
16
|
Zhang Q, Liu Z, Huang X, Heng X, Wu J, Chen Z, Guo X, Fan J, Huang Q. MDIVI-1 ALLEVIATES SEPSIS-INDUCED LIVER INJURY BY INHIBITING STING SIGNALING ACTIVATION. Shock 2024; 62:95-102. [PMID: 38526162 DOI: 10.1097/shk.0000000000002349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
ABSTRACT Proinflammatory hyperactivation of Kupffer cells (KCs) is foremost involved in the pathogenesis of sepsis-induced liver injury. Our previous study found that stimulator of interferon genes (STING) signaling was activated in KCs in response of lipopolysaccharide (LPS) and knocking down dynamin-related protein 1 (DRP1) in KCs effectively inhibited the activation of STING signaling and the subsequent production of proinflammatory cytokines. In this study, we demonstrated that in vivo treatment with mitochondrial division inhibitor 1 (Mdivi-1), a selective inhibitor of DRP1, alleviated cecal ligation and puncture (CLP)-induced liver injury with the improvement of liver pathology and function. Moreover, we found that STING in liver was mainly concentrated in KCs and STING signaling was significantly activated in KCs after CLP. The STING deficiency effectively ameliorated liver injury and decreased the mortality of septic mice, which were reversely worsened by the enhanced activation of STING with DMXAA. The further study showed that Mdivi-1 markedly attenuated STING signaling activation in KCs and inhibited systemic inflammatory response. Importantly, DMXAA application in CLP mice blunted Mdivi-1's liver protection effect. Taken together, our study confirmed Mdivi-1 effectively alleviated CLP-induced liver injury partially through inhibiting STING signaling activation in KCs, which provides new insights and a novel potential pharmacological therapeutic target for treating septic liver injury.
Collapse
Affiliation(s)
| | - Zhuanhua Liu
- Guangdong Provincial Key Lab of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaoxia Huang
- Guangdong Provincial Key Lab of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xin Heng
- Guangdong Provincial Key Lab of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jie Wu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhenfeng Chen
- Guangdong Provincial Key Lab of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaohua Guo
- Guangdong Provincial Key Lab of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jun Fan
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | | |
Collapse
|
17
|
Tan N, Jian G, Peng J, Tian X, Chen B. Chishao - Fuzi herbal pair restore the macrophage M1/M2 balance in acute-on-chronic liver failure. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118010. [PMID: 38499260 DOI: 10.1016/j.jep.2024.118010] [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: 10/25/2023] [Revised: 02/21/2024] [Accepted: 03/03/2024] [Indexed: 03/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional herbal pair Paeoniae Radix Rubra (roots of Paeonia lactiflora Pall., Chishao in Chinese) and Aconiti Lateralis Radix Praeparata (lateral roots of Aconitum carmichaelii Debeaux, Fuzi in Chinese) are widely used for the treatment of liver diseases, demonstrating clinical efficacy against acute-on-chronic liver failure (ACLF). As the core drug pair representing the "clearing method" and "warming method" in traditional Chinese medicine (TCM), they align with the TCM syndromic characteristics of ACLF, characterized by a mixture of deficiencies and realities. However, the molecular mechanisms underlying the anti-ACLF effects of Chishao - Fuzi herbal pair remain unclear. AIM OF THE STUDY To reveal the immunoinflammatory status of patients with hepatitis B virus-related ACLF (HBV-ACLF) based on macrophage polarization and to explore the mechanism of action of Chishao - Fuzi herbal pair in regulating macrophage polarization against ACLF. MATERIALS AND METHODS Peripheral blood samples were prospectively obtained from patients with HBV-ACLF, patients with chronic hepatitis B (CHB) in the immunoactive phase and healthy individuals. Flow cytometry, qRT-qPCR, and ELISA were used to reveal the activation status of monocyte-macrophages and the expression differences in related cytokines in the peripheral blood of patients with HBV-ACLF. Then, an ACLF rat model and a macrophage inflammation model in vitro were established. Hematoxylin-eosin staining, immunohistochemical staining, transmission electron microscopy, flow cytometry, western blotting, RT-qPCR, and ELISA were used to observe changes in the expression of M1/M2 macrophage markers and related inflammatory factors after Chishao - Fuzi herbal pair intervention, both in vivo and in vitro. RESULTS Patients with HBV-ACLF exhibited an imbalance in M1/M2 macrophage polarization, showing a tendency to activate M1 macrophages with high expression of CD86 and iNOS. This imbalance led to an increase in relevant pro-inflammatory factors (IL-1β, IL-6, TNF-α) and a decrease in anti-inflammatory factors (IL-10, TGF-β, VEGF), exacerbating the uncontrolled immune-inflammatory response. Chishao - Fuzi herbal pair intervention improved liver function, coagulation function, and histopathological injury in ACLF rats. It also partially ameliorated endotoxemia and inflammatory injury in ACLF. The mechanism was to restore the immune-inflammatory imbalance and prevent the exacerbation of inflammatory response to liver failure by promoting macrophage polarization toward M2 anti-inflammatory direction, inhibiting M1 macrophage activation, and increasing the levels of anti-inflammatory factors and decreasing pro-inflammatory factors. CONCLUSION Chishao - Fuzi herbal pair can reduce the systemic inflammatory burden of liver failure by modulating macrophage polarization and restoring ACLF immune-inflammatory imbalance. This study provides new perspectives and strategies for studying HBV-ACLF immune reconstitution and inflammatory response control.
Collapse
Affiliation(s)
- Nianhua Tan
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China; Department of Hepatology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan Province, China.
| | - Gonghui Jian
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Jie Peng
- Department of Hepatology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan Province, China
| | - Xuefei Tian
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China; Hunan Province University Key Laboratory of Oncology of Traditional Chinese Medicine Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China; Key Laboratory of Traditional Chinese Medicine for Mechanism of Tumor Prevention &Treatment, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| | - Bin Chen
- Department of Hepatology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan Province, China.
| |
Collapse
|
18
|
Ding X, Pang Y, Liu Q, Zhang H, Wu J, Lei J, Zhang T. GO-PEG Represses the Progression of Liver Inflammation via Regulating the M1/M2 Polarization of Kupffer Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306483. [PMID: 38229561 DOI: 10.1002/smll.202306483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 01/03/2024] [Indexed: 01/18/2024]
Abstract
As a highly promising nanomaterial, exploring the impact of the liver, a vital organ, stands out as a crucial focus in the examination of its biological effects. Kupffer cells (KCs) are one of the first immune cells to contact with exotic-substances in liver. Therefore, this study investigates the immunomodulatory effects and mechanisms of polyethylene glycol-modified graphene oxide (GO-PEG) on KCs. Initial RNA-seq and KEGG pathway analyses reveal the inhibition of the TOLL-like receptor, TNF-α and NOD-like receptor pathways in continually stimulated KCs exposed to GO-PEG. Subsequent biological experiments validate that a 48-hour exposure to GO-PEG alleviates LPS-induced KCs immune activation, characterized by a shift in polarization from M1 to M2. The underlying mechanism involves the absorption of double-stranded RNA/single-stranded RNA, inhibiting the activation of TLR3 and TLR7 in KCs. Employing a Kupffer/AML12 cell co-culture model and animal studies, it is observed that GO-PEG indirectly inhibit oxidative stress, mitochondrial dysfunction, and apoptosis in AML12 cells, partially mitigating systemic inflammation and preserving liver tissue/function. This effect is attributed to the paracrine interaction between KCs and hepatocytes. These findings suggest a meaningful and effective strategy for treating liver inflammation, particularly when combined with anti-inflammatory drugs.
Collapse
Affiliation(s)
- Xiaomeng Ding
- Ministry of Education Key Laboratory of Environmental Medicine Engineering, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yanting Pang
- Ministry of Education Key Laboratory of Environmental Medicine Engineering, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Qing Liu
- Ministry of Education Key Laboratory of Environmental Medicine Engineering, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Haopeng Zhang
- Ministry of Education Key Laboratory of Environmental Medicine Engineering, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Jiawei Wu
- Ministry of Education Key Laboratory of Environmental Medicine Engineering, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Jialin Lei
- Ministry of Education Key Laboratory of Environmental Medicine Engineering, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Ting Zhang
- Ministry of Education Key Laboratory of Environmental Medicine Engineering, School of Public Health, Southeast University, Nanjing, 210009, China
- Jiangsu key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210009, China
| |
Collapse
|
19
|
Lei Y, Yu H, Ding S, Liu H, Liu C, Fu R. Molecular mechanism of ATF6 in unfolded protein response and its role in disease. Heliyon 2024; 10:e25937. [PMID: 38434326 PMCID: PMC10907738 DOI: 10.1016/j.heliyon.2024.e25937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/04/2024] [Accepted: 02/05/2024] [Indexed: 03/05/2024] Open
Abstract
Activating transcription factor 6 (ATF6), an important signaling molecule in unfolded protein response (UPR), plays a role in the pathogenesis of several diseases, including diseases such as congenital retinal disease, liver fibrosis and ankylosing spondylitis. After endoplasmic reticulum stress (ERS), ATF6 is activated after separation from binding immunoglobulin protein (GRP78/BiP) in the endoplasmic reticulum (ER) and transported to the Golgi apparatus to be hydrolyzed by site 1 and site 2 proteases into ATF6 fragments, which localize to the nucleus and regulate the transcription and expression of ERS-related genes. In these diseases, ERS leads to the activation of UPR, which ultimately lead to the occurrence and development of diseases by regulating the physiological state of cells through the ATF6 signaling pathway. Here, we discuss the evidence for the pathogenic importance of ATF6 signaling in different diseases and discuss preclinical results.
Collapse
Affiliation(s)
| | | | - Shaoxue Ding
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Chunyan Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| |
Collapse
|
20
|
Márton RA, Sebők C, Mackei M, Tráj P, Vörösházi J, Kemény Á, Neogrády Z, Mátis G. Cecropin A: investigation of a host defense peptide with multifaceted immunomodulatory activity in a chicken hepatic cell culture. Front Vet Sci 2024; 11:1337677. [PMID: 38496311 PMCID: PMC10940386 DOI: 10.3389/fvets.2024.1337677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/20/2024] [Indexed: 03/19/2024] Open
Abstract
Introduction Host defense peptides (HDPs) are increasingly referred to as promising candidates for the reduction of the use of conventional antibiotics, thereby combating antibiotic resistance. As HDPs have been described to exert various immunomodulatory effects, cecropin A (CecA) appears to be a potent agent to influence the host inflammatory response. Methods In the present study, a chicken primary hepatocyte-non-parenchymal cell co-culture was used to investigate the putative immunomodulatory effects of CecA alone and in inflammatory conditions evoked by polyinosinic-polycytidylic acid (Poly I:C). To examine the viability of the cells, the extracellular lactate dehydrogenase (LDH) activity was determined by colorimetric assay. Inflammatory markers interleukin (IL)-8 and transforming growth factor-ß1 (TGF-ß1) were investigated using the ELISA method, whereas concentrations of IL-6, IL-10, and interferon-γ (IFN-γ) were assayed by Luminex xMAP technology. Extracellular H2O2 and malondialdehyde levels were measured by fluorometric and colorimetric methods, respectively. Results Results of the lower concentrations suggested the safe application of CecA; however, it might contribute to hepatic cell membrane damage at its higher concentrations. We also found that the peptide alleviated the inflammatory response, reflected by the decreased production of the pro-inflammatory IL-6, IL-8, and IFN-γ. In addition, CecA diminished the levels of anti-inflammatory IL-10 and TGF-ß1. The oxidative markers measured remained unchanged in most cases of CecA exposure. Discussion CecA displayed a multifaceted immunomodulatory but not purely anti-inflammatory activity on the hepatic cells, and might be suggested to maintain the hepatic inflammatory homeostasis in Poly I:C-triggered immune response. To conclude, our study suggests that CecA might be a promising molecule for the development of new immunomodulatory antibiotic-substitutive agents in poultry medicine; however, there is still a lot to clarify regarding its cellular effects.
Collapse
Affiliation(s)
- Rege Anna Márton
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, Budapest, Hungary
| | - Csilla Sebők
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
| | - Máté Mackei
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, Budapest, Hungary
| | - Patrik Tráj
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
| | - Júlia Vörösházi
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
| | - Ágnes Kemény
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Department of Medical Biology, Medical School, University of Pécs, Pécs, Hungary
| | - Zsuzsanna Neogrády
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
| | - Gábor Mátis
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, Budapest, Hungary
| |
Collapse
|
21
|
Yang Y, Zhang JY, Ma ZJ, Wang SC, He P, Tang XQ, Yang CF, Luo X, Yang X, Li L, Zhang MC, Li Y, Yu JH. Visualization of therapeutic intervention for acute liver injury using low-intensity pulsed ultrasound-responsive phase variant nanoparticles. Biomater Sci 2024; 12:1281-1293. [PMID: 38252410 DOI: 10.1039/d3bm01423a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Acute liver injury (ALI) is a highly fatal condition characterized by sudden massive necrosis of liver cells, inflammation, and impaired coagulation function. Currently, the primary clinical approach for managing ALI involves symptom management based on the underlying causes. The association between excessive reactive oxygen species originating from macrophages and acute liver injury is noteworthy. Therefore, we designed a novel nanoscale phase variant contrast agent, denoted as PFP@CeO2@Lips, which effectively scavenges reactive oxygen species, and enables visualization through low intensity pulsed ultrasound activation. The efficacy of the nanoparticles in scavenging excess reactive oxygen species from RAW264.7 and protective AML12 cells has been demonstrated through in vitro and in vivo experiments. Additionally, these nanoparticles have shown a protective effect against LPS/D-GalN attack in C57BL/6J mice. Furthermore, when exposed to LIPUS irritation, the nanoparticles undergo liquid-gas phase transition and enable ultrasound imaging.
Collapse
Affiliation(s)
- You Yang
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Ju-Ying Zhang
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Zi-Jun Ma
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Shi-Chun Wang
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Ping He
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Xiao-Qing Tang
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Chao-Feng Yang
- Department of Radiology, Affifiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Xia Luo
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Xing Yang
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Ling Li
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Mao-Chun Zhang
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Yang Li
- Department of Ultrasound, Yuechi People's Hospital, Guangan, 638300, Sichuan, China
- Department of Radiology, Affifiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Jin-Hong Yu
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
- Department of Ultrasound, Yuechi People's Hospital, Guangan, 638300, Sichuan, China
| |
Collapse
|
22
|
Yang Y, Tian T, Wang Z, Li S, Li N, Luo H, Jiang Y. LncRNA 220, a newly discovered long non-conding RNA inhibiting apoptosis and autophagy in Kupffer cells in LPS-induced endotoxemic mice through the XBP1u-PI3K-AKT pathway. Int Immunopharmacol 2024; 128:111497. [PMID: 38241842 DOI: 10.1016/j.intimp.2024.111497] [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: 10/20/2023] [Revised: 12/20/2023] [Accepted: 01/03/2024] [Indexed: 01/21/2024]
Abstract
Sepsis is recognized as a potentially fatal condition characterized by acute organ dysfunction resulting from an imbalanced immune response to infection. Acute liver injury (ALI) arises as an inflammatory outcome of immune response dysregulation associated with sepsis. Kupffer cells, which are liver-specific macrophages, are known to have a significant impact on ALI, although the precise regulatory mechanism remains unclear. Numerous studies have showcased the regulatory impact of long non-coding RNAs (lncRNAs) on the progression of diverse ailments, yet their precise regulatory mechanisms remain predominantly unexplored. In this study, a novel long non-coding RNA (lncRNA), referred to as lncRNA 220, was discovered using high-throughput sequencing. The expression of lncRNA 220 was found to be significantly elevated in the livers of mice with lipopolysaccharide (LPS)-induced endotoxemia, specifically during the 8-hour time period. Furthermore, in Kupffer cells treated with LPS, lncRNA 220 was observed to inhibit apoptosis and autophagy by activating the PI3K-AKT-mTORC1 pathway. This effect was achieved through the reduction of X-box protein 1 unspliced (Xbp1u) mRNA stability and suppression of its translation in the context of endoplasmic reticulum stress (ERS). Ultimately, this intervention mitigated the progression of LPS-induced ALI. To summarize, our study establishes lncRNA 220 as a newly identified regulator that suppresses apoptosis and autophagy in Kupffer cells subjected to LPS treatment, indicating its potential as a molecular target for ALI in endotoxemic mice.
Collapse
Affiliation(s)
- Ying Yang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tian Tian
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhenqi Wang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shan Li
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Nanhong Li
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Haihua Luo
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Respiratory and Critical Care Medicine, Affiliated Dongguan Hospital, Southern Medical University, Guangdong, China.
| |
Collapse
|
23
|
Li Q, Zhang F, Wang H, Tong Y, Fu Y, Wu K, Li J, Wang C, Wang Z, Jia Y, Chen R, Wu Y, Cui R, Wu Y, Qi Y, Qu K, Liu C, Zhang J. NEDD4 lactylation promotes APAP induced liver injury through Caspase11 dependent non-canonical pyroptosis. Int J Biol Sci 2024; 20:1413-1435. [PMID: 38385085 PMCID: PMC10878146 DOI: 10.7150/ijbs.91284] [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: 10/19/2023] [Accepted: 01/07/2024] [Indexed: 02/23/2024] Open
Abstract
Caspase-11 detection of intracellular lipopolysaccharide mediates non-canonical pyroptosis, which could result in inflammatory damage and organ lesions in various diseases such as sepsis. Our research found that lactate from the microenvironment of acetaminophen-induced acute liver injury increased Caspase-11 levels, enhanced gasdermin D activation and accelerated macrophage pyroptosis, which lead to exacerbation of liver injury. Further experiments unveiled that lactate inhibits Caspase-11 ubiquitination by reducing its binding to NEDD4, a negative regulator of Caspase-11. We also identified that lactates regulated NEDD4 K33 lactylation, which inhibits protein interactions between Caspase-11 and NEDD4. Moreover, restraining lactylation reduces non-canonical pyroptosis in macrophages and ameliorates liver injury. Our work links lactate to the exquisite regulation of the non-canonical inflammasome, and provides a basis for targeting lactylation signaling to combat Caspase-11-mediated non-canonical pyroptosis and acetaminophen-induced liver injury.
Collapse
Affiliation(s)
- Qinglin Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of Vascular Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Fengping Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Hai Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Yingmu Tong
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
| | - Yunong Fu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Kunjin Wu
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, People's Republic of China
| | - Jing Li
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, People's Republic of China
| | - Cong Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Zi Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Yifan Jia
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
| | - Rui Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Yang Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Ruixia Cui
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Yi Wu
- MOE Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
| | - Yun Qi
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
| | - Kai Qu
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, People's Republic of China
| | - Chang Liu
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, People's Republic of China
| | - Jingyao Zhang
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of SICU, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
| |
Collapse
|
24
|
Wu KC, Adedeji AO, Zabka TS, Hosseini I, Kenkre R, Getz JA, Nguyen T, Decalf J, Bainbridge TW, Chilton JA, Moussion CC, Rao GK. Nonclinical pharmacokinetics, pharmacodynamics and safety assessment of a FLT3L-Fc molecule for cancer immunotherapy. Toxicol Appl Pharmacol 2024; 483:116837. [PMID: 38278496 DOI: 10.1016/j.taap.2024.116837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/14/2024] [Accepted: 01/23/2024] [Indexed: 01/28/2024]
Abstract
FLT3L-Fc is a cytokine-Fc fusion agonizing receptor-type tyrosine-protein kinase FLT3 (fms-related tyrosine kinase 3; CD135). FLT3 is expressed on dendritic cells (DCs) as well as myeloid and lymphoid progenitors. Nonclinical pharmacokinetics, pharmacodynamics and safety of FLT3L-Fc were investigated in rats and cynomolgus monkeys. FLT3L-Fc induced robust pharmacodynamic responses, evidenced by marked expansion of peripheral blood cDC1s, cDC2s, and pDCs (up to 301-fold in rats and 378-fold in monkeys), peaking at 8-10 days after the first dose. FLT3L-Fc was well tolerated with no adverse findings at doses up to 10 mg/kg administered intravenously twice three weeks apart. In both species, major clinical pathology findings consisted of expansion of white blood cell (WBC) populations including lymphocytes, monocytes, neutrophils, basophils, and large unstained cells, which were pronounced after the first dose. The WBC findings were associated microscopically with histiocytic and mononuclear cell infiltrates in multiple organs. Tissue immunohistochemistry in monkeys showed that the leukocyte infiltrates consisted of hematopoietic progenitor cells and histiocytes with a reactive morphology and were associated with a slight stimulation of regional T and B cell populations. Additional FLT3L-Fc-associated changes included decreases in red blood cell (RBC) mass, increases in RBC distribution width, variable changes in reticulocytes, and transient alterations in platelet counts (rats only). The RBC and WBC findings were associated microscopically with increased hematopoietic cellularity of the bone marrow in both species and increased splenic megakaryocytic extramedullary hematopoiesis in rats. The totality of nonclinical safety data support the clinical development of FLT3L-Fc.
Collapse
Affiliation(s)
- Kai Connie Wu
- Department of Safety Assessment, Genentech Inc., South San Francisco, CA 94080, United States of America
| | - Adeyemi O Adedeji
- Department of Safety Assessment, Genentech Inc., South San Francisco, CA 94080, United States of America
| | - Tanja S Zabka
- Department of Safety Assessment, Genentech Inc., South San Francisco, CA 94080, United States of America
| | - Iraj Hosseini
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., South San Francisco, CA 94080, United States of America
| | - Radhika Kenkre
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., South San Francisco, CA 94080, United States of America
| | - Jennifer A Getz
- Department of Bioanalytical Sciences, Genentech Inc., South San Francisco, CA 94080, United States of America
| | - Tien Nguyen
- Department of Safety Assessment, Genentech Inc., South San Francisco, CA 94080, United States of America
| | - Jérémie Decalf
- Department of Cancer Immunology, Genentech Inc., South San Francisco, CA 94080, United States of America
| | - Travis W Bainbridge
- Department of Protein Chemistry, Genentech Inc., South San Francisco, CA 94080, United States of America
| | | | - Christine C Moussion
- Department of Cancer Immunology, Genentech Inc., South San Francisco, CA 94080, United States of America
| | - Gautham K Rao
- Department of Safety Assessment, Genentech Inc., South San Francisco, CA 94080, United States of America.
| |
Collapse
|
25
|
Puengel T, Tacke F. Role of Kupffer cells and other immune cells. SINUSOIDAL CELLS IN LIVER DISEASES 2024:483-511. [DOI: 10.1016/b978-0-323-95262-0.00024-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
26
|
Li H, Yu S, Liu H, Chen L, Liu H, Liu X, Shen C. Immunologic barriers in liver transplantation: a single-cell analysis of the role of mesenchymal stem cells. Front Immunol 2023; 14:1274982. [PMID: 38143768 PMCID: PMC10748593 DOI: 10.3389/fimmu.2023.1274982] [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: 08/09/2023] [Accepted: 11/13/2023] [Indexed: 12/26/2023] Open
Abstract
Background This study aimed to analyze the biomarkers that may reliably indicate rejection or tolerance and the mechanism that underlie the induction and maintenance of liver transplantation (LT) tolerance related to immunosuppressant or mesenchymal stem cells (MSCs). Methods LT models of Lewis-Lewis and F344-Lewis rats were established. Lewis-Lewis rats model served as a control (Syn). F344-Lewis rats were treated with immunosuppressant alone (Allo+IS) or in combination with MSCs (Allo+IS+MSCs). Intrahepatic cell composition particularly immune cells was compared between the groups by single-cell sequencing. Analysis of subclusters, KEGG pathway analysis, and pseudotime trajectory analysis were performed to explore the potential immunoregulatory mechanisms of immunosuppressant alone or combined with MSCs. Results Immunosuppressants alone or combined with MSCs increases the liver tolerance, to a certain extent. Single-cell sequencing identified intrahepatic cell composition signature, including cell subpopulations of B cells, cholangiocytes, endothelial cells, erythrocytes, hepatic stellate cells, hepatocytes, mononuclear phagocytes, neutrophils, T cells, and plasmacytoid dendritic cells. Immunosuppressant particularly its combination with MSCs altered the landscape of intrahepatic cells in transplanted livers, as well as gene expression patterns in immune cells. MSCs may be included in the differentiation of T cells, classical monocytes, and non-classical monocytes. Conclusion These findings provided novel insights for better understanding the heterogeneity and biological functions of intrahepatic immune cells after LT treated by IS alone or in combination with MSCs. The identified markers of immune cells may serve as the immunotherapeutic targets for MSC treatment of liver transplant rejection.
Collapse
Affiliation(s)
- Haitao Li
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Saihua Yu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Haiyan Liu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Lihong Chen
- Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Hongzhi Liu
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Xingwen Liu
- Department of Nursing, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Conglong Shen
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
27
|
Li L, Lan Y, Wang F, Gao T. Linarin Protects Against CCl 4-Induced Acute Liver Injury via Activating Autophagy and Inhibiting the Inflammatory Response: Involving the TLR4/MAPK/Nrf2 Pathway. Drug Des Devel Ther 2023; 17:3589-3604. [PMID: 38076631 PMCID: PMC10700044 DOI: 10.2147/dddt.s433591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023] Open
Abstract
Background Linarin has been implicated in the inhibition of inflammatory responses and hepatoprotective effects. However, the precise mechanism by which Linarin integrates injury-induced signaling from inflammatory responses and oxidative stress remains unclear. Methods We evaluated the role of Linarin in a mouse model of carbon tetrachloride (CCl4)-induced acute liver injury. Mice were orally pretreated with Linarin or vehicle for seven consecutive days, followed by intraperitoneal injection with 0.2% (v/v) CCl4. To investigate the mechanism of action on oxidative stress, CCl4-stimulated HepG2 cells were utilized. Results Our results revealed Linarin remarkably attenuated the loss of hepatic architecture, inflammatory cell infiltration, serum transaminases, and pro-inflammatory cytokines induced by CCl4. Linarin attenuated CCl4-induced oxidative stress by increasing the expression of cytosolic Nrf2 (nuclear factor erythroid 2-related factor 2), inducing nuclear localization of Nrf2, and increasing stress-induced protein heme oxygenase-1 (HO-1). Additionally, Linarin decreased the expression of toll-like receptors (TLR)-4, and its downstream proteins, MyD88, IRAK1, and TRAF6. Furthermore, Linarin reversed CCl4-induced phosphorylation of ERK, p38, and JNK. Importantly, Linarin increased the expression of both LC3II and Beclin 1, which are hallmarks of autophagic flux. Autophagy-mediated hepatoprotective effects in Linarin-treated HepG2 cells were mitigated by the autophagy inhibitor 3-MA. However, combined treatment of Linarin with 3-MA failed to significantly reverse cell apoptosis and the production of transaminases and pro-inflammatory cytokines. Conclusion Linarin prevents acute liver injury, possibly by alleviating ROS-induced oxidative stress, inhibiting TLR4/MyD88 and JNK/p38/ERK-mediated inflammatory responses, and promoting Beclin 1/LC3II-mediated autophagic flux.
Collapse
Affiliation(s)
- Lulu Li
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, People’s Republic of China
- Department of Pharmacy, Wuhan NO.1 Hospital, Wuhan, Hubei, People’s Republic of China
| | - Yan Lan
- Department of Pharmacy, Huangshi Central Hospital, Huangshi, Hubei, People’s Republic of China
| | - Fuqian Wang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, People’s Republic of China
- Department of Pharmacy, Wuhan NO.1 Hospital, Wuhan, Hubei, People’s Republic of China
| | - Tiexiang Gao
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, People’s Republic of China
| |
Collapse
|
28
|
Li LX, Wang L, Wang S, Zhang XN, Liu H, Zhang YJ, Wu CT, Zhang CL, Zeng T. Allyl methyl disulfide (AMDS) prevents N,N-dimethyl formamide-induced liver damage by suppressing oxidative stress and NLRP3 inflammasome activation. Food Chem Toxicol 2023; 182:114198. [PMID: 37995826 DOI: 10.1016/j.fct.2023.114198] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 10/30/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
N,N-dimethylformamide (DMF), a widely consumed industrial solvent with persistent characteristics, can induce occupational liver damage and pose threats to the general population due to the enormous DMF-containing industrial efflux and emission from indoor facilities. This study was performed to explore the roles of allyl methyl disulfide (AMDS) in liver damage induced by DMF and the underlying mechanisms. AMDS was found to effectively suppress the elevation in the liver weight/body weight ratio and serum aminotransferase activities, and reduce the mortality of mice induced by DMF. In addition, AMDS abrogated DMF-elicited increases in malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE) levels and decreases in glutathione (GSH) levels in mouse livers. The increase in macrophage number, mRNA expression of M1 macrophage biomarkers, and protein expression of key components in the NF-κB pathway and NLRP3 inflammasome induced by DMF exposure were all suppressed by AMDS in mouse livers. Furthermore, AMDS inhibited DMF-induced cell damage and NF-κB activation in cocultured AML12 hepatocytes and J774A.1 macrophages. However, AMDS per se did not significantly affect the protein level and activity of CYP2E1. Collectively, these results demonstrate that AMDS effectively ameliorates DMF-induced acute liver damage possibly by suppressing oxidative stress and inactivating the NF-κB pathway and NLRP3 inflammasome.
Collapse
Affiliation(s)
- Long-Xia Li
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lin Wang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Shuo Wang
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong, 252059, China
| | - Xiu-Ning Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Hong Liu
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yan-Jing Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Chuan-Tao Wu
- The Animal Research Center, Shandong University, Jinan, Shandong, 250012, China
| | - Cui-Li Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
29
|
Xie D, Ouyang S. The role and mechanisms of macrophage polarization and hepatocyte pyroptosis in acute liver failure. Front Immunol 2023; 14:1279264. [PMID: 37954583 PMCID: PMC10639160 DOI: 10.3389/fimmu.2023.1279264] [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: 08/17/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
Acute liver failure (ALF) is a severe liver disease caused by disruptions in the body's immune microenvironment. In the early stages of ALF, Kupffer cells (KCs) become depleted and recruit monocytes derived from the bone marrow or abdomen to replace the depleted macrophages entering the liver. These monocytes differentiate into mature macrophages, which are activated in the immune microenvironment of the liver and polarized to perform various functions. Macrophage polarization can occur in two directions: pro-inflammatory M1 macrophages and anti-inflammatory M2 macrophages. Controlling the ratio and direction of M1 and M2 in ALF can help reduce liver injury. However, the liver damage caused by pyroptosis should not be underestimated, as it is a caspase-dependent form of cell death. Inhibiting pyroptosis has been shown to effectively reduce liver damage induced by ALF. Furthermore, macrophage polarization and pyroptosis share common binding sites, signaling pathways, and outcomes. In the review, we describe the role of macrophage polarization and pyroptosis in the pathogenesis of ALF. Additionally, we preliminarily explore the relationship between macrophage polarization and pyroptosis, as well as their effects on ALF.
Collapse
Affiliation(s)
| | - Shi Ouyang
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, Department of Infectious Diseases, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
30
|
Lee JL, Wang YC, Hsu YA, Chen CS, Weng RC, Lu YP, Chuang CY, Wan L. Galectin-12 modulates Kupffer cell polarization to alter the progression of nonalcoholic fatty liver disease. Glycobiology 2023; 33:673-682. [PMID: 37504513 DOI: 10.1093/glycob/cwad062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/22/2023] [Accepted: 07/16/2023] [Indexed: 07/29/2023] Open
Abstract
Nonalcoholic fatty liver disease is caused by an imbalance in lipid metabolism and immune response to pose a risk factor for liver fibrosis. Recent evidence indicates that M2 macrophages secrete transforming growth factor-β1, which contributes to liver fibrosis. Galectin-12 has been demonstrated to regulate lipid metabolism and macrophage polarization. The purpose of this study is to investigate the role of galectin-12 in the development of nonalcoholic fatty liver disease and fibrosis. Liver tissue from wild-type C57BL/6 mice fed with a high-fat diet containing cholesterol and cholic acid for 4-12 weeks was used to examine galectin-12 expression and its correlation with nonalcoholic fatty liver disease. Furthermore, the effects of galectin-12 on M2 macrophages during the progression of nonalcoholic fatty liver disease were investigated by studying Kupffer cells from galectin-12 knockout mice and doxycycline-inducible Gal12-/-THP-1 cells. Ablation of galectin-12 promoted M2 polarization of Kupffer cells, as indicated by higher levels of M2 markers, such as arginase I and chitinase 3-like protein 3. Furthermore, the activation of signal transducer and activator of transcription 6 was significantly higher in Gal12-/- macrophages activated by interleukin-4, which was correlated with higher levels of transforming growth factor-β1. Moreover, Gal12-/- macrophage-conditioned medium promoted hepatic stellate cells myofibroblast differentiation, which was indicated by higher α-smooth muscle actin expression levels compared with those treated with LacZ control medium. Finally, we demonstrated that galectin-12 knockdown negatively regulated the suppressor of cytokine signaling 3 levels. These findings suggested that galectin-12 balances M1/M2 polarization of Kupffer cells to prevent nonalcoholic fatty liver disease progression.
Collapse
Affiliation(s)
- Jyun-Lin Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yao-Chien Wang
- Department of Emergency Medicine, Taichung Tzu Chi Hospital, Taichung 427, Taiwan
| | - Yu-An Hsu
- School of Chinese Medicine, China Medical University, Taichung 404, Taiwan
| | - Chih-Sheng Chen
- Division of Chinese Medicine, Asia University Hospital, Taichung 413, Taiwan
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung 401, Taiwan
- Department of Chinese Medicine, China Medicine University Hospital, Taichung 404, Taiwan
| | - Rui-Cian Weng
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 106, Taiwan
- National Applied Research Laboratories, Taiwan Instrument Research Institute, Hsinchu 300, Taiwan
| | - Yen-Pei Lu
- National Applied Research Laboratories, Taiwan Instrument Research Institute, Hsinchu 300, Taiwan
| | - Chun-Yu Chuang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Lei Wan
- School of Chinese Medicine, China Medical University, Taichung 404, Taiwan
- Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung 404, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 413, Taiwan
| |
Collapse
|
31
|
Kandhi R, Yeganeh M, Yoshimura A, Menendez A, Ramanathan S, Ilangumaran S. Hepatic stellate cell-intrinsic role of SOCS1 in controlling hepatic fibrogenic response and the pro-inflammatory macrophage compartment during liver fibrosis. Front Immunol 2023; 14:1259246. [PMID: 37860002 PMCID: PMC10582746 DOI: 10.3389/fimmu.2023.1259246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
Introduction Hepatic stellate cells (HSC) become activated, differentiate to myofibroblasts and produce extracellular fibrillar matrix during liver fibrosis. The hepatic fibrogenic response is orchestrated by reciprocal interactions between HSCs and macrophages and their secreted products. SOCS1 can regulate several cytokines and growth factors implicated in liver fibrosis. Here we investigated the role of SOCS1 in regulating HSC activation. Methods Mice lacking SOCS1 in HSCs (Socs1ΔHSC) were generated by crossing Socs1fl/fl and LratCre mice. Liver fibrosis was induced by carbon tetrachloride and evaluated by Sirius red staining, hydroxyproline content and immunostaining of myofibroblasts. Gene expression of pro-fibrogenic factors, cytokines, growth factors and chemokines were quantified by RT-qPCR. The phenotype and the numbers of intrahepatic leukocyte subsets were studied by flow cytometry. The impact of fibrosis on the development of diethyl nitrosamine-induced hepatocellular carcinoma was evaluated. Results Socs1ΔHSC mice developed more severe liver fibrosis than control Socs1fl/fl mice that was characterized by increased collagen deposition and myofibroblast differentiation. Socs1ΔHSC mice showed a significant increase in the expression of smooth muscle actin, collagens, matrix metalloproteases, cytokines, growth factors and chemokines in the liver following fibrosis induction. The fibrotic livers of Socs1ΔHSC mice displayed heightened inflammatory cell infiltration with increased proportion and numbers of Ly6ChiCCR2+ pro-inflammatory macrophages. This macrophage population contained elevated numbers of CCR2+CX3CR1+ cells, suggesting impaired transition towards restorative macrophages. Fibrosis induction following exposure to diethyl nitrosamine resulted in more numerous and larger liver tumor nodules in Socs1ΔHSC mice than in Socs1fl/fl mice. Discussion Our findings indicate that (i) SOCS1 expression in HSCs is a critical to control liver fibrosis and development of hepatocaellular carcinoma, and (ii) attenuation of HSC activation by SOCS1 regulates pro-inflammatory macrophage recruitment and differentiation during liver fibrosis.
Collapse
Affiliation(s)
- Rajani Kandhi
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mehdi Yeganeh
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Alfredo Menendez
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Sheela Ramanathan
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Subburaj Ilangumaran
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
32
|
Yang Y, Jin Y, Zhu X, Rao Q, Zhao Z, Yang J. Hepatotoxicity evaluation and possible mechanisms of decabrominated diphenyl ethers (BDE-209) in broilers: Oxidative stress, inflammatory, and transcriptomics. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115460. [PMID: 37696076 DOI: 10.1016/j.ecoenv.2023.115460] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/25/2023] [Accepted: 09/07/2023] [Indexed: 09/13/2023]
Abstract
Decabrominated diphenyl ether (BDE-209), a persistent organic pollutant, is linked to a great number of health problems, the most severe of which impact the liver due to its role in the elimination and degradation of exogenous harmful substances. Though the hepatotoxicity of BDE-209 has been observed, its underlying mechanism is yet unknown. The purpose of this study is to thoroughly investigate the hepatotoxicity of BDE-209 and its molecular processes in broilers by subjecting 120 male broilers to varied concentrations of BDE-209 for 42 days. We observed that the bioaccumulation of BDE-209 in the liver in a dose-dependent manner, and that BDE-209 exposure can raise the concentrations of ALT, AST, and GGT, accompanied by hepatocyte fatty degeneration and inflammatory foci. In the hepatic homogenates, oxidative stress was evidenced by elevated levels of MDA and ROS and decreased activies of SOD and CAT. Additionally, pro-inflammatory cytokines including IL-1, IL-1β, TNF-α, IL-8 levels were increased, whereas anti-inflammatory cytokine IL-4 level was declined. Furthermore, RNA sequencing revealed that genes involved in inflammation were considerably dysregulated, and real-time PCR verified the expressed alterations of numerous genes related to the MAPK and WNT signaling pathways. The protein concentrations of NF-κB, β-catenin, and WNT5A, and the phosphorylation levels of JNK and ERK were all dramatically enhanced. The current study indicates that BDE-209 exposure can cause hepatotoxicity in broilers via bioaccumulation and oxidative stress, which then activates the MAPK and WNT signaling pathways, subsequently generating inflammation and hepatic injury.
Collapse
Affiliation(s)
- Yi Yang
- Institute for Agri-Food Standard and Testing, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Yuhong Jin
- College of Food Science and Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Xing Zhu
- College of Animal Science, Guizhou University, Guiyang, Guizhou 550025, China
| | - Qinxiong Rao
- Institute for Agri-Food Standard and Testing, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Zhihui Zhao
- Institute for Agri-Food Standard and Testing, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Junhua Yang
- Institute for Agri-Food Standard and Testing, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China.
| |
Collapse
|
33
|
Heo MJ, Suh JH, Poulsen KL, Ju C, Kim KH. Updates on the Immune Cell Basis of Hepatic Ischemia-Reperfusion Injury. Mol Cells 2023; 46:527-534. [PMID: 37691258 PMCID: PMC10495686 DOI: 10.14348/molcells.2023.0099] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 06/19/2023] [Accepted: 07/21/2023] [Indexed: 09/12/2023] Open
Abstract
Liver ischemia-reperfusion injury (IRI) is the main cause of organ dysfunction and failure after liver surgeries including organ transplantation. The mechanism of liver IRI is complex and numerous signals are involved but cellular metabolic disturbances, oxidative stress, and inflammation are considered the major contributors to liver IRI. In addition, the activation of inflammatory signals exacerbates liver IRI by recruiting macrophages, dendritic cells, and neutrophils, and activating NK cells, NKT cells, and cytotoxic T cells. Technological advances enable us to understand the role of specific immune cells during liver IRI. Accordingly, therapeutic strategies to prevent or treat liver IRI have been proposed but no definitive and effective therapies exist yet. This review summarizes the current update on the immune cell functions and discusses therapeutic potentials in liver IRI. A better understanding of this complex and highly dynamic process may allow for the development of innovative therapeutic approaches and optimize patient outcomes.
Collapse
Affiliation(s)
- Mi Jeong Heo
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ji Ho Suh
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Kyle L. Poulsen
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Cynthia Ju
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Kang Ho Kim
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
34
|
Hassan GS, Flores Molina M, Shoukry NH. The multifaceted role of macrophages during acute liver injury. Front Immunol 2023; 14:1237042. [PMID: 37736102 PMCID: PMC10510203 DOI: 10.3389/fimmu.2023.1237042] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/15/2023] [Indexed: 09/23/2023] Open
Abstract
The liver is situated at the interface of the gut and circulation where it acts as a filter for blood-borne and gut-derived microbes and biological molecules, promoting tolerance of non-invasive antigens while driving immune responses against pathogenic ones. Liver resident immune cells such as Kupffer cells (KCs), a subset of macrophages, maintain homeostasis under physiological conditions. However, upon liver injury, these cells and others recruited from circulation participate in the response to injury and the repair of tissue damage. Such response is thus spatially and temporally regulated and implicates interconnected cells of immune and non-immune nature. This review will describe the hepatic immune environment during acute liver injury and the subsequent wound healing process. In its early stages, the wound healing immune response involves a necroinflammatory process characterized by partial depletion of resident KCs and lymphocytes and a significant infiltration of myeloid cells including monocyte-derived macrophages (MoMFs) complemented by a wave of pro-inflammatory mediators. The subsequent repair stage includes restoring KCs, initiating angiogenesis, renewing extracellular matrix and enhancing proliferation/activation of resident parenchymal and mesenchymal cells. This review will focus on the multifaceted role of hepatic macrophages, including KCs and MoMFs, and their spatial distribution and roles during acute liver injury.
Collapse
Affiliation(s)
- Ghada S. Hassan
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Manuel Flores Molina
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Naglaa H. Shoukry
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
35
|
Novi S, Vestuto V, Campiglia P, Tecce N, Bertamino A, Tecce MF. Anti-Angiogenic Effects of Natural Compounds in Diet-Associated Hepatic Inflammation. Nutrients 2023; 15:2748. [PMID: 37375652 DOI: 10.3390/nu15122748] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Alcoholic liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD) are the most common causes of chronic liver disease and are increasingly emerging as a global health problem. Such disorders can lead to liver damage, resulting in the release of pro-inflammatory cytokines and the activation of infiltrating immune cells. These are some of the common features of ALD progression in ASH (alcoholic steatohepatitis) and NAFLD to NASH (non-alcoholic steatohepatitis). Hepatic steatosis, followed by fibrosis, lead to a continuous progression accompanied by angiogenesis. This process creates hypoxia, which activates vascular factors, initiating pathological angiogenesis and further fibrosis. This forms a vicious cycle of ongoing damage and progression. This condition further exacerbates liver injury and may contribute to the development of comorbidities, such as metabolic syndrome as well as hepatocellular carcinoma. Increasing evidence suggests that anti-angiogenic therapy may have beneficial effects on these hepatic disorders and their exacerbation. Therefore, there is a great interest to deepen the knowledge of the molecular mechanisms of natural anti-angiogenic products that could both prevent and control liver diseases. In this review, we focus on the role of major natural anti-angiogenic compounds against steatohepatitis and determine their potential therapeutic benefits in the treatment of liver inflammation caused by an imbalanced diet.
Collapse
Affiliation(s)
- Sara Novi
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy
| | - Nicola Tecce
- Unit of Endocrinology, Department of Clinical Medicine and Surgery, Medical School of Naples, Federico II University, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy
| | - Mario Felice Tecce
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy
| |
Collapse
|
36
|
Attia AA, Hamad HA, Fawzy MA, Saleh SR. The Prophylactic Effect of Vitamin C and Vitamin B12 against Ultraviolet-C-Induced Hepatotoxicity in Male Rats. Molecules 2023; 28:molecules28114302. [PMID: 37298780 DOI: 10.3390/molecules28114302] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Ultraviolet C (UVC) devices are an effective means of disinfecting surfaces and protecting medical tools against various microbes, including coronavirus. Overexposure to UVC can induce oxidative stress, damage the genetic material, and harm biological systems. This study investigated the prophylactic efficacy of vitamin C and B12 against hepatotoxicity in UVC-intoxicated rats. Rats were irradiated with UVC (725.76, 967.68, and 1048.36 J/cm2) for 2 weeks. The rats were pretreated with the aforementioned antioxidants for two months before UVC irradiation. The prophylactic effect of vitamins against UVC hepatotoxicity was evaluated by monitoring the alteration of liver enzyme activities, antioxidant status, apoptotic and inflammatory markers, DNA fragmentation, and histological and ultrastructural alterations. Rats exposed to UVC showed a significant increase in liver enzymes, oxidant-antioxidant balance disruption, and increased hepatic inflammatory markers (TNF-α, IL-1β, iNOS, and IDO-1). Additionally, obvious over-expression of activated caspase-3 protein and DNA fragmentation were detected. Histological and ultrastructural examinations verified the biochemical findings. Co-treatment with vitamins ameliorated the deviated parameters to variable degrees. In conclusion, vitamin C could alleviate UVC-induced hepatotoxicity more than vitamin B12 by diminishing oxidative stress, inflammation, and DNA damage. This study could provide a reference for the clinical practice of vitamin C and B12 as radioprotective for workers in UVC disinfectant areas.
Collapse
Affiliation(s)
- Azza A Attia
- Zoology Department, Faculty of Science, Alexandria University, Alexandria 21515, Egypt
| | - Huda A Hamad
- Zoology Department, Faculty of Science, Alexandria University, Alexandria 21515, Egypt
- Zoology Department, Faculty of Science, Omar Al-Mukhtar University, Al Bayda 00218, Libya
| | - M Adel Fawzy
- Physics Department, Faculty of Science, Alexandria University, Alexandria 21515, Egypt
| | - Samar R Saleh
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21515, Egypt
| |
Collapse
|
37
|
Xu GX, Wei S, Yu C, Zhao SQ, Yang WJ, Feng YH, Pan C, Yang KX, Ma Y. Activation of Kupffer cells in NAFLD and NASH: mechanisms and therapeutic interventions. Front Cell Dev Biol 2023; 11:1199519. [PMID: 37261074 PMCID: PMC10228659 DOI: 10.3389/fcell.2023.1199519] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/05/2023] [Indexed: 06/02/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are emerging as the leading causes of liver disease worldwide. These conditions can lead to cirrhosis, liver cancer, liver failure, and other related ailments. At present, liver transplantation remains the sole treatment option for end-stage NASH, leading to a rapidly growing socioeconomic burden. Kupffer cells (KCs) are a dominant population of macrophages that reside in the liver, playing a crucial role in innate immunity. Their primary function includes phagocytosing exogenous substances, presenting antigens, and triggering immune responses. Moreover, they interact with other liver cells during the pathogenesis of NAFLD, and this crosstalk may either delay or exacerbate disease progression. Stimulation by endogenous signals triggers the activation of KCs, resulting in the expression of various inflammatory factors and chemokines, such as NLRP3, TNF-α, IL-1B, and IL-6, and contributing to the inflammatory cascade. In the past 5 years, significant advances have been made in understanding the biological properties and immune functions of KCs in NAFLD, including their interactions with tissue molecules, underlying molecular mechanisms, signaling pathways, and relevant therapeutic interventions. Having a comprehensive understanding of these mechanisms and characteristics can have enormous potential in guiding future strategies for the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yong Ma
- *Correspondence: Kun-Xing Yang, ; Yong Ma,
| |
Collapse
|
38
|
Ge J, Li H, Yang JQ, Yue Y, Lu SY, Nie HY, Zhang T, Sun PM, Yan HF, Sun HW, Yang JW, Zhou JL, Cui Y. Autophagy in hepatic macrophages can be regulator and potential therapeutic target of liver diseases: A review. Medicine (Baltimore) 2023; 102:e33698. [PMID: 37171337 PMCID: PMC10174421 DOI: 10.1097/md.0000000000033698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/06/2023] [Accepted: 04/14/2023] [Indexed: 05/13/2023] Open
Abstract
Hepatic macrophages are a complex population of cells that play an important role in the normal functioning of the liver and in liver diseases. Autophagy, as a maintainer of cellular homeostasis, is closely connected to many liver diseases. And its roles are not always beneficial, but manifesting as a double-edged sword. The polarization of macrophages and the activation of inflammasomes are mediated by intracellular and extracellular signals, respectively, and are important ways for macrophages to take part in a variety of liver diseases. More attention should be paid to autophagy of hepatic macrophages in liver diseases. In this review, we focus on the regulatory role of hepatic macrophages' autophagy in a variety of liver diseases; especially on the upstream regulator of polarization and inflammasomes activation of the hepatic macrophages. We believe that the autophagy of hepatic macrophages can become a potential therapeutic target for management of liver diseases.
Collapse
Affiliation(s)
- Jun Ge
- Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
- Department of General Surgery, The 306th Hospital of PLA-Peking University Teaching Hospital, Beijing 100101, China
| | - Hao Li
- Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Jia-Qi Yang
- Department of General Surgery, The 306th Hospital of PLA-Peking University Teaching Hospital, Beijing 100101, China
| | - Yuan Yue
- Department of General Surgery, The 306th Hospital of PLA-Peking University Teaching Hospital, Beijing 100101, China
| | - Sheng-Yu Lu
- Department of General Surgery, The 306th Hospital of PLA-Peking University Teaching Hospital, Beijing 100101, China
| | - Hong-Yun Nie
- Department of General Surgery, The 306th Hospital of PLA-Peking University Teaching Hospital, Beijing 100101, China
| | - Tao Zhang
- Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Pei-Ming Sun
- Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Hong-Feng Yan
- Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Hong-Wei Sun
- Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Jian-Wu Yang
- Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Jin-Lian Zhou
- Department of Pathology, Strategic Support Force Medical Center, Beijing 100101, China
| | - Yan Cui
- Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
- Department of General Surgery, The 306th Hospital of PLA-Peking University Teaching Hospital, Beijing 100101, China
| |
Collapse
|
39
|
Ito Y, Hosono K, Amano H. Responses of hepatic sinusoidal cells to liver ischemia–reperfusion injury. Front Cell Dev Biol 2023; 11:1171317. [PMID: 37082623 PMCID: PMC10112669 DOI: 10.3389/fcell.2023.1171317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 03/27/2023] [Indexed: 04/07/2023] Open
Abstract
The liver displays a remarkable regenerative capacity in response to acute liver injury. In addition to the proliferation of hepatocytes during liver regeneration, non-parenchymal cells, including liver macrophages, liver sinusoidal endothelial cells (LSECs), and hepatic stellate cells (HSCs) play critical roles in liver repair and regeneration. Liver ischemia–reperfusion injury (IRI) is a major cause of increased liver damage during liver resection, transplantation, and trauma. Impaired liver repair increases postoperative morbidity and mortality of patients who underwent liver surgery. Successful liver repair and regeneration after liver IRI requires coordinated interplay and synergic actions between hepatic resident cells and recruited cell components. However, the underlying mechanisms of liver repair after liver IRI are not well understood. Recent technological advances have revealed the heterogeneity of each liver cell component in the steady state and diseased livers. In this review, we describe the progress in the biology of liver non-parenchymal cells obtained from novel technological advances. We address the functional role of each cell component in response to liver IRI and the interactions between diverse immune repertoires and non-hematopoietic cell populations during the course of liver repair after liver IRI. We also discuss how these findings can help in the design of novel therapeutic approaches. Growing insights into the cellular interactions during liver IRI would enhance the pathology of liver IRI understanding comprehensively and further develop the strategies for improvement of liver repair.
Collapse
|
40
|
Tong H, Wang L, Zhang K, Shi J, Wu Y, Bao Y, Wang C. S100A6 Activates Kupffer Cells via the p-P38 and p-JNK Pathways to Induce Inflammation, Mononuclear/macrophage Infiltration Sterile Liver Injury in Mice. Inflammation 2023; 46:534-554. [PMID: 36484925 DOI: 10.1007/s10753-022-01750-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/17/2022] [Accepted: 09/30/2022] [Indexed: 12/13/2022]
Abstract
Noninfectious liver injury, including the effects of chemical material, drugs and diet, is a major cause of liver diseases worldwide. In chemical and drugs-induced liver injury, innate inflammatory responses are mediated by extracellular danger signals. The S100 protein can act as danger signals, which can promote the migration and chemotaxis of immune cells, promote the release of various inflammatory cytokines, and regulate the body's inflammatory and immune responses. However, the role of S100A6 in inflammatory response in chemical and drugs-induced sterile liver injury remains unclear. We constructed the model of sterile liver injury induced by carbon tetrachloride (CCl4)/Paracetamol (APAP) and performed RNA sequencing (RNA-seq) on the liver tissues after injury (days 2 and 5). We analyzed inflammatory protein secretion in the liver tissue supernatant by enzyme-linked immunosorbent assay (ELISA), determined the inflammation response by bioinformatic analysis during sterile liver injury, and assessed mononuclear/macrophage infiltration by immunohistochemistry and flow cytometry. Immunohistochemistry was used to analyze the location of S100A6. We conducted inflammatory factor expression analysis and molecular mechanistic studies in Kupffer cells (KCs) induced by S100A6 using quantitative reverse transcription-polymerase chain reaction (qRT-PCR), ELISA, and western blot in vitro experiments. We performed chemokine CCL2 expression analysis and molecular mechanism studies using the same method. We used a Transwell assay to show the infiltration of mononuclear/macrophage. We here observed that aggravated inflammatory response was shown in CCl4 and APAP-administrated mice, as evidenced by enhanced production of inflammatory cytokines (TNF-α, IL-1β), and elevated mononuclear/macrophage infiltration and activation of immunity. The expression of S100A6 was significantly increased on day 2 after sterile liver injury, which is primarily produced by injured liver cells. Mechanistic studies established that S100A6 activates Kupffer cells (KCs) via the p-P38, p-JNK and P65 pathways to induce inflammation in vitro. Furthermore, TNF-α can stimulate liver cells via the p-P38 and p-JNK pathways to produce CCL2 and promote the infiltration of mononuclear/macrophage. In summary, we showed that S100A6 plays an important role in regulating inflammation, thus influencing sterile liver injury. Our findings provide novel evidence that S100A6 can as a danger signal that contributes to pro-inflammatory activation through p-P38 and p-JNK pathways in CCl4 and APAP-induced sterile liver injury in mice. In addition, the inflammatory factor TNF-α induces a large amount of CCL2 production in normal liver cells surrounding the injured area through a paracrine action, which is chemotactic for blood mononuclear/macrophage infiltration.
Collapse
Affiliation(s)
- He Tong
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Li Wang
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, Inner Mongolia, China
| | - Kefan Zhang
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Jing Shi
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Yongshuai Wu
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Yulong Bao
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, Inner Mongolia, China.
| | - Changshan Wang
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China.
| |
Collapse
|
41
|
Ma J, Xu Y, Zhang M, Li Y. Geraniol ameliorates acute liver failure induced by lipopolysaccharide/D-galactosamine via regulating macrophage polarization and NLRP3 inflammasome activation by PPAR-γ methylation Geraniol alleviates acute liver failure. Biochem Pharmacol 2023; 210:115467. [PMID: 36849063 DOI: 10.1016/j.bcp.2023.115467] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/18/2023] [Accepted: 02/21/2023] [Indexed: 03/01/2023]
Abstract
Geraniol (Ger), a natural acyclic monoterpene alcohol, has been reported to exert protective effects through anti-inflammation in Acute liver failure (ALF). However, its specific roles and precise mechanisms underlying anti-inflammatory effects in ALF have not yet fully explored. We aimed to investigated the hepatoprotective effects and mechanisms of Ger against ALF induced by lipopolysaccharide (LPS)/D-galactosamine (GaIN). In this study, the liver tissue and serum of LPS/D-GaIN-induced mice were collected. The degree of liver tissue injury was evaluated by HE and TUNEL staining. Serum levels of liver injury markers (ALT and AST) and inflammatory factors were measured by ELISA assays. PCR and western blotting were conducted to determine the expression of inflammatory cytokines, NLRP3 inflammasome-related proteins, PPAR-γ pathway-related proteins, DNA Methyltransferases and M1/M2 polarization cytokines. Immunofluorescence staining was used to assess the localization and expression of macrophage markers (F4/80 and CD86), NLRP3 and PPAR-γ. In vitro experiments were performed in macrophages stimulated with LPS with or without IFN-γ. Purification of macrophages and cell apoptosis was analyzed using flow cytometry. We found that Ger effectively alleviated ALF in mice, specified by the attenuation of liver tissue pathological damage, inhibition of ALT, AST and inflammatory factor levels, and inactivation of NLRP3 inflammasome. Meanwhile, downregulation M1 macrophage polarization may involve in the protective effects of Ger. In vitro, Ger reduced the activation of NLRP3 inflammasome and apoptosis through regulating PPAR-γ methylation by inhibiting M1 macrophage polarization. In conclusion, Ger protects against ALF through suppressing NLRP3 inflammasome-mediated inflammation and LPS-induced macrophage M1 polarization via modulating PPAR-γ methylation.
Collapse
Affiliation(s)
- Jing Ma
- Infectious Disease Department, The Second XIANGYA Hospital of Central South University, Changsha, Hunan, China
| | - Yun Xu
- Infectious Disease Department, The Second XIANGYA Hospital of Central South University, Changsha, Hunan, China
| | - Min Zhang
- Infectious Disease Department, The Second XIANGYA Hospital of Central South University, Changsha, Hunan, China
| | - Yi Li
- Infectious Disease Department, The Second XIANGYA Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
42
|
Liu Y, Sheng M, Jia L, Zhu M, Yu W. Protective effects of cordycepin pretreatment against liver ischemia/reperfusion injury in mice. Immun Inflamm Dis 2023; 11:e792. [PMID: 36988254 PMCID: PMC10013135 DOI: 10.1002/iid3.792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/20/2023] [Accepted: 02/09/2023] [Indexed: 03/16/2023] Open
Abstract
INTRODUCTION Cordycepin has been reported to exhibit hepatic protective and anti-inflammatory properties. Here, we investigated the role of cordycepin in ischemia/reperfusion (IR)-induced liver injury in a mouse model. METHODS Mice were pretreated with cordycepin by gavage for 3 weeks, followed by the establishment of the IR modeling. Liver injury, Suzuki's histological grading, hepatic apoptosis, and inflammatory responses were evaluated by biochemical and pathological analysis. RESULTS It was found that Cordycepin pretreatment at 50 mg/kg for 3 weeks attenuated IR-induced liver injury, as reflected by the significant decrease of the levels of aspartate aminotransferase, alanine transaminase, lactate dehydrogenase, and low-density lipoprotein. Cordycepin pretreatment also reduced histopathological changes, attenuated hepatocyte apoptosis, inflammatory responses in the livers of IR mice. Mechanically, toll-like receptor 4/nuclear factor kappa-B signaling in liver tissues was inhibited by Cordycepin pretreatment. CONCLUSIONS In conclusion, Cordycepin pretreatment protects IR-induced liver injury, which demonstrates its potential for the treatment of IR in the liver.
Collapse
Affiliation(s)
- Yunxia Liu
- Department of AnesthesiologyTianjin First Central HospitalTianjinChina
| | - Mingwei Sheng
- Department of AnesthesiologyTianjin First Central HospitalTianjinChina
| | - Lili Jia
- Department of AnesthesiologyTianjin First Central HospitalTianjinChina
| | - Min Zhu
- Department of AnesthesiologyTianjin First Central HospitalTianjinChina
| | - Wenli Yu
- Department of AnesthesiologyTianjin First Central HospitalTianjinChina
| |
Collapse
|
43
|
Xu L, Wang H. A dual role of inflammation in acetaminophen-induced liver injury. LIVER RESEARCH 2023; 7:9-15. [PMID: 39959696 PMCID: PMC11791818 DOI: 10.1016/j.livres.2023.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/14/2023] [Accepted: 03/05/2023] [Indexed: 03/12/2023]
Abstract
In many affluent nations, acetaminophen (APAP) overdose is the leading cause of drug-induced acute liver failure. The process of APAP-induced liver injury (AILI) is intimately tied to inflammation, including hepatocyte necrosis-caused initiation of inflammation, inflammation amplification that exacerbates liver injury, and the resolution of inflammation that triggers liver regeneration and repair. Excessive APAP metabolism in the liver eventually leads to hepatocyte necrosis and inflammation. Innate immune cells, such as neutrophils, eosinophils, monocytes, and gammadelta T cells, are recruited into the injured liver and release various cytokines. These immune cells and cytokines have been found to serve two purposes in AILI. In this review, we highlighted the dual role of inflammation, including inflammatory cytokines and inflammatory immune cells in AILI, and discussed possible explanations for contradictory findings.
Collapse
Affiliation(s)
- Long Xu
- School of Basic Medical Science, Anhui Medical University, Hefei, Anhui, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
44
|
The effect of miR-155-5p on M1 polarization of Kupffer cells and immune response during liver transplantation through regulating the expression of KDM5D. Mol Immunol 2023; 155:17-26. [PMID: 36641950 DOI: 10.1016/j.molimm.2023.01.003] [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: 10/16/2022] [Revised: 12/12/2022] [Accepted: 01/06/2023] [Indexed: 01/15/2023]
Abstract
BACKGROUND To explore the effect and its specific mechanism of miR-155-5p on M1 polarization of Kupffer cells (KCs) and immune response in liver transplantation (LT) through KDM5D. METHODS Primary KCs were isolated from Wistar rats and identified by cell culture, ink-swallowing test and flow cytometry. The cells identified as KCs were induced into LT acute rejection (AR) model cells by LPS/IFN-γ, flow cytometry was used for cell sorting and apoptosis detection. Enzyme-linked immunosorbent assay (ELISA) kit was used to detect the levels of inflammatory factors, macrophages and liver function markers. RT-qPCR detected the expression of miR-155-5p and KDM5D mRNA. The protein expression of KDM5D was detected by Western blot. Dual luciferase reporter gene experiment verified the targeting relationship between miR-155-5p and KDM5D. RESULTS The separated KCs adhered after being cultured for 24 h, had pseudopodia and phagocytosis, and the proportion of F4/80 positive cells was more than 90%. The expression of miR-155-5p was increased in LPS/IFN-γ-induced KCs. And knockdown of miR-155-5p inhibited H3K4me3 and H3K27me3 of TNF-α promoter, M1 polarization of KCs and the immune response of AR model cells by upregulating KDM5D. In animal experiments, knockdown of miR-155-5p was found to inhibit liver damage and immune response in rats with allogeneic orthotopic LT. CONCLUSION These results confirmed that miR-155-5p inhibited M1 polarization of KCs induced by LPS/IFN-γ, thereby alleviating AR and liver function impairment after LT by upregulating KDM5D.
Collapse
|
45
|
Li H, Hu P, Zou Y, Yuan L, Xu Y, Zhang X, Luo X, Zhang Z. Tanshinone IIA and hepatocellular carcinoma: A potential therapeutic drug. Front Oncol 2023; 13:1071415. [PMID: 36798821 PMCID: PMC9928209 DOI: 10.3389/fonc.2023.1071415] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 01/04/2023] [Indexed: 02/04/2023] Open
Abstract
Because of its high prevalence and poor long-term clinical treatment effect, liver disease is regarded as a major public health problem around the world. Among them, viral hepatitis, fatty liver, cirrhosis, non-alcoholic fatty liver disease (NAFLD), and autoimmune liver disease are common causes and inducements of liver injury, and play an important role in the occurrence and development of hepatocellular carcinoma (HCC). Tanshinone IIA (TsIIA) is a fat soluble polyphenol of Salvia miltiorrhiza that is extracted from Salvia miltiorrhiza. Because of its strong biological activity (anti-inflammatory, antioxidant), it is widely used in Asia to treat cardiovascular and liver diseases. In addition, TsIIA has shown significant anti-HCC activity in previous studies. It not only has significant anti proliferation and pro apoptotic properties. It can also play an anti-cancer role by mediating a variety of signal pathways, including phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/rapamycin (mTOR), mitogen-activated protein kinase (MAPK), and nuclear factor kappa-B (NF-κB). This review not only reviews the existing evidence and molecular mechanism of TsIIA's anti-HCC effect but also reviews the liver-protective effect of TsIIA and its impact on liver fibrosis, NAFLD, and other risk factors for liver cancer. In addition, we also conducted network pharmacological analysis on TsIIA and HCC to further screen and explore the possible targets of TsIIA against hepatocellular carcinoma. It is expected to provide a theoretical basis for the development of anti-HCC-related drugs based on TsIIA.
Collapse
Affiliation(s)
- Hu Li
- Emergency Department, Affiliated Hospital of Binzhou Medical College, Binzhou, China
| | - Pengbo Hu
- Emergency Department, Affiliated Hospital of Binzhou Medical College, Binzhou, China,Institute of Medical Science of Binzhou Medical University, Yantai, China
| | - Yajun Zou
- Emergency Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lijuan Yuan
- Emergency Department, Affiliated Hospital of Binzhou Medical College, Binzhou, China
| | - Yucheng Xu
- Emergency Department, Affiliated Hospital of Binzhou Medical College, Binzhou, China
| | - Xiaohui Zhang
- Emergency Department, Affiliated Hospital of Binzhou Medical College, Binzhou, China
| | - Xiaoyan Luo
- Emergency Department, Affiliated Hospital of Binzhou Medical College, Binzhou, China
| | - Zhiqiang Zhang
- Emergency Department, Affiliated Hospital of Binzhou Medical College, Binzhou, China,Institute of Medical Science of Binzhou Medical University, Yantai, China,*Correspondence: Zhiqiang Zhang,
| |
Collapse
|
46
|
Ren M, Zhang J, Dai S, Wang C, Chen Z, Zhang S, Xu J, Qin X, Liu F. CX3CR1 deficiency exacerbates immune-mediated hepatitis by increasing NF-κB-mediated cytokine production in macrophage and T cell. Exp Biol Med (Maywood) 2023; 248:117-129. [PMID: 36426712 PMCID: PMC10041049 DOI: 10.1177/15353702221128573] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Immune-mediated hepatitis is marked by liver inflammation characterized by immune cell infiltration, chemokine/cytokine production, and hepatocyte injury. C-X3C motif receptor 1 (CX3CR1), as the receptor of chemokine C-X3C motif ligand 1 (CX3CL1)/fractalkine, is mainly expressed on immune cells including monocytes and T cells. Previous studies have shown that CX3CR1 protects against liver fibrosis, but the exact role of CX3CL1/CX3CR1 in acute immune-mediated hepatitis remains unknown. Here, we investigate the role of the CX3CL1/CX3CR1 axis in immune-mediated hepatitis using concanavalin A (ConA)-induced liver injury model in CX3CR1-deficient (Cx3cr1-/-) mice. We observed that Cx3cr1-/- mice had severe liver injury and increased pro-inflammatory cytokines (tumor necrosis factor-alpha [TNF-α], interferon-gamma [IFN-γ], interleukin-1 beta [IL-1β], and IL-6) in serum and liver compared to wild-type (Cx3cr1+/+) mice after ConA injection. The deficiency of CX3CR1 did not affect ConA-induced immune cell infiltration in liver but led to elevated production of TNF-α in macrophages as well as IFN-γ in T cells after ConA treatment. On the contrary, exogenous CX3CL1 attenuated ConA-induced cytokine production in wild type, but not CX3CR1-deficient macrophages and T cells. Furthermore, in vitro results showed that CX3CR1 deficiency promoted the pro-inflammatory cytokine expression by increasing the phosphorylation of nuclear factor kappa B (NF-κB) p65 (p-NF-κB p65). Finally, pre-treatment of p-NF-κB p65 inhibitor, resveratrol, attenuated ConA-induced liver injury and inflammatory responses, especially in Cx3cr1-/- mice. In conclusion, our data show that the deficiency of CX3CR1 promotes pro-inflammatory cytokine production in macrophages and T cells by enhancing the phosphorylation of NF-κB p65, which exacerbates liver injury in ConA-induced hepatitis.
Collapse
Affiliation(s)
- Mi Ren
- Department of Hepatobiliary Surgery and Liver Transplantation, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Jinyan Zhang
- Department of Hepatobiliary Surgery and Liver Transplantation, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Shen Dai
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250000, People's Republic of China
| | - Chenxiao Wang
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Zheng Chen
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Siqi Zhang
- Department of Hepatobiliary Surgery and Liver Transplantation, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Junming Xu
- Department of Hepatobiliary Surgery and Liver Transplantation, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Xuebin Qin
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Fengming Liu
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250000, People's Republic of China
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
47
|
Khodayari N, Oshins R, Aranyos AM, Duarte S, Mostofizadeh S, Lu Y, Brantly M. Characterization of hepatic inflammatory changes in a C57BL/6J mouse model of alpha1-antitrypsin deficiency. Am J Physiol Gastrointest Liver Physiol 2022; 323:G594-G608. [PMID: 36256438 DOI: 10.1152/ajpgi.00207.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Alpha-1 antitrypsin deficiency (AATD) is a genetic disease caused by a hepatic accumulation of mutant alpha-1 antitrypsin (ZAAT). Individuals with AATD are prone to develop a chronic liver disease that remains undiagnosed until late stage of the disease. Here, we sought to characterize the liver pathophysiology of a human transgenic mouse model for AATD with a manifestation of liver disease compared with normal transgenic mice model. Male and female transgenic mice for normal (Pi*M) and mutant variant (Pi*Z) human alpha-1 antitrypsin at 3 and 6 mo of age were subjected to this study. The progression of hepatic ZAAT accumulation, hepatocyte injury, steatosis, liver inflammation, and fibrotic features were monitored by performing an in vivo study. We have also performed a Next-Gene transcriptomic analysis of the transgenic mice liver tissue 16 h after lipopolysaccharide (LPS) administration to delineate liver inflammatory response in Pi*Z mice as compared with Pi*M. Our results show hepatic ZAAT accumulation, followed by hepatocyte ballooning and liver steatosis developed at 3 mo in Pi*Z mice compared with the mice carrying normal variant of human alpha-1 antitrypsin. We observed higher levels of hepatic immune cell infiltrations in both 3- and 6-mo-old Pi*Z mice compared with Pi*M as an indication of liver inflammation. Liver fibrosis was observed as accumulation of collagen in 6-mo-old Pi*Z liver tissues compared with Pi*M control mice. Furthermore, the transcriptomic analysis revealed a dysregulated liver immune response to LPS in Pi*Z mice compared with Pi*M. Of particular interest for translational work, this study aims to establish a mouse model of AATD with a strong manifestation of liver disease that will be a valuable in vivo tool to study the pathophysiology of AATD-mediated liver disease. Our data suggest that the human transgenic mouse model of AATD could provide a suitable model for the evaluation of therapeutic approaches and preventive reagents against AATD-mediated liver disease.NEW & NOTEWORTHY We have characterized a mouse model of human alpha-1 antitrypsin deficiency with a strong manifestation of liver disease that can be used as an in vivo tool to test preventive and therapeutic reagents. Our data explores the altered immunophenotype of alpha-1 antitrypsin-deficient liver macrophages and suggests a relationship between acute inflammation, immune response, and fibrosis.
Collapse
Affiliation(s)
- Nazli Khodayari
- Division of Pulmonary, Department of Medicine, University of Florida, Gainesville, Florida
| | - Regina Oshins
- Division of Pulmonary, Department of Medicine, University of Florida, Gainesville, Florida
| | - Alek M Aranyos
- Division of Pulmonary, Department of Medicine, University of Florida, Gainesville, Florida
| | - Sergio Duarte
- Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida
| | - Sayedamin Mostofizadeh
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Yuanqing Lu
- Division of Pulmonary, Department of Medicine, University of Florida, Gainesville, Florida
| | - Mark Brantly
- Division of Pulmonary, Department of Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
48
|
Li Q, Chen F, Wang F. The immunological mechanisms and therapeutic potential in drug-induced liver injury: lessons learned from acetaminophen hepatotoxicity. Cell Biosci 2022; 12:187. [PMID: 36414987 PMCID: PMC9682794 DOI: 10.1186/s13578-022-00921-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/01/2022] [Indexed: 11/24/2022] Open
Abstract
Acute liver failure caused by drug overdose is a significant clinical problem in developed countries. Acetaminophen (APAP), a widely used analgesic and antipyretic drug, but its overdose can cause acute liver failure. In addition to APAP-induced direct hepatotoxicity, the intracellular signaling mechanisms of APAP-induced liver injury (AILI) including metabolic activation, mitochondrial oxidant stress and proinflammatory response further affect progression and severity of AILI. Liver inflammation is a result of multiple interactions of cell death molecules, immune cell-derived cytokines and chemokines, as well as damaged cell-released signals which orchestrate hepatic immune cell infiltration. The immunoregulatory interplay of these inflammatory mediators and switching of immune responses during AILI lead to different fate of liver pathology. Thus, better understanding the complex interplay of immune cell subsets in experimental models and defining their functional involvement in disease progression are essential to identify novel therapeutic targets for the treatment of AILI. Here, this present review aims to systematically elaborate on the underlying immunological mechanisms of AILI, its relevance to immune cells and their effector molecules, and briefly discuss great therapeutic potential based on inflammatory mediators.
Collapse
Affiliation(s)
- Qianhui Li
- grid.511083.e0000 0004 7671 2506Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, No.628, Zhenyuan Road, Shenzhen, 518107 China
| | - Feng Chen
- grid.511083.e0000 0004 7671 2506Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, No.628, Zhenyuan Road, Shenzhen, 518107 China
| | - Fei Wang
- grid.511083.e0000 0004 7671 2506Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, No.628, Zhenyuan Road, Shenzhen, 518107 China
| |
Collapse
|
49
|
Zhao Q, Sheng MF, Wang YY, Wang XY, Liu WY, Zhang YY, Ke TY, Chen S, Pang GZ, Yong L, Ding Z, Shen YJ, Shen YX, Shao W. LncRNA Gm26917 regulates inflammatory response in macrophages by enhancing Annexin A1 ubiquitination in LPS-induced acute liver injury. Front Pharmacol 2022; 13:975250. [PMID: 36386180 PMCID: PMC9663662 DOI: 10.3389/fphar.2022.975250] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 10/20/2022] [Indexed: 09/08/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are defined as transcripts of more than 200 nucleotides that have little or no coding potential. LncRNAs function as key regulators in diverse physiological and pathological processes. However, the roles of lncRNAs in lipopolysaccharide (LPS)-induced acute liver injury (ALI) are still elusive. In this study, we report the roles of lncRNA Gm26917 induced by LPS in modulating liver inflammation. As key components of the innate immune system, macrophages play critical roles in the initiation, progression and resolution of ALI. Our studies demonstrated that Gm26917 localized in the cytoplasm of hepatic macrophages and globally regulated the expression of inflammatory genes and the differentiation of macrophages. In vivo study showed that lentivirus-mediated gene silencing of Gm26917 attenuated liver inflammation and protected mice from LPS-induced ALI. Furthermore, mechanistic study showed that the 3'-truncation of Gm26917 interacted with the N-terminus of Annexin A1, a negative regulator of the NF-κB signaling pathway. We also found that Gm26917 knockdown suppressed NF-κB activity by decreasing the ubiquitination of Annexin A1 and its interaction with NEMO. In addition, expression of Gm26917 in inflammatory macrophages was regulated by the transcription factor forkhead box M1 (FOXM1). LPS treatment dramatically increased the binding of FOXM1 to the promoter region of Gm26917 in macrophages. In summary, our findings suggest that lncRNA Gm26917 silencing protects against LPS-induced liver injury by regulating the TLR4/NF-κB signaling pathway in macrophages.
Collapse
Affiliation(s)
- Qing Zhao
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, Anhui, China
| | - Meng-Fei Sheng
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, Anhui, China
| | - Yao-Yun Wang
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, Anhui, China
| | - Xing-Yu Wang
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, Anhui, China
| | - Wei-Yi Liu
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, Anhui, China
| | - Yuan-Yuan Zhang
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, Anhui, China
| | - Tiao-Ying Ke
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, Anhui, China
| | - Shu Chen
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, Anhui, China
| | - Gao-Zong Pang
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, Anhui, China
| | - Liang Yong
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Zhan Ding
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Science, Wuhan University, Wuhan, Hubei, China
| | - Yu-Jun Shen
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, Anhui, China
| | - Yu-Xian Shen
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, Anhui, China
| | - Wei Shao
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
50
|
Hong J, Kim Y. Fatty Liver/Adipose Tissue Dual-Targeting Nanoparticles with Heme Oxygenase-1 Inducer for Amelioration of Obesity, Obesity-Induced Type 2 Diabetes, and Steatohepatitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203286. [PMID: 36209391 PMCID: PMC9685446 DOI: 10.1002/advs.202203286] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/15/2022] [Indexed: 05/28/2023]
Abstract
Persistent uptake of high-calorie diets induces the storage of excessive lipid in visceral adipose tissue. Lipids secreted from obese adipose tissue are accumulated in peripheral tissues such as the liver, pancreas, and muscle, and impair insulin sensitivity causing type 2 diabetes mellitus (T2DM). Furthermore, the accumulation of inflammatory cytokines and lipids in the liver induces apoptosis and fibrogenesis, and ultimately causes nonalcoholic steatohepatitis (NASH). To modulate obese tissue environments, it is challenged to selectively deliver inducers of heme oxygenase-1 (HO-1) to adipose tissue with the aid of a prohibitin targeting drug delivery system. Prohibitin binding peptide (PBP), an oligopeptide targeting prohibitin rich in adipose tissue, is conjugated on the surface of Hemin- or CoPP-loaded poly(lactide-co-glycolide) nanoparticles (PBP-NPs). PBP-NPs efficiently differentiate lipid storing white adipocytes into energy-generating brown adipocytes in T2DM and NASH models. In addition, PBP-NPs are found to target prohibitin overexpressed fatty liver in the NASH model and inhibit hepatic uptake of circulating lipids. Furthermore, PBP-NPs switch phenotypes of inflammatory macrophages in damaged organs and lower inflammation. Taken together, dual-targeted induction of HO-1 in fatty adipose and liver tissues is proven to be a promising therapeutic strategy to ameliorate obesity, insulin resistance, and steatohepatitis by lowering lipids and cytokines.
Collapse
Affiliation(s)
- Juhyeong Hong
- Department of BioengineeringInstitute for Bioengineering and Biopharmaceutical Research Hanyang UniversitySeoul04763South Korea
- Education and Research Group for Biopharmaceutical Innovation LeaderHanyang UniversitySeoul04763South Korea
| | - Yong‐Hee Kim
- Department of BioengineeringInstitute for Bioengineering and Biopharmaceutical Research Hanyang UniversitySeoul04763South Korea
- Education and Research Group for Biopharmaceutical Innovation LeaderHanyang UniversitySeoul04763South Korea
| |
Collapse
|