1
|
Zhang J, Wang Y, Fan M, Guan Y, Zhang W, Huang F, Zhang Z, Li X, Yuan B, Liu W, Geng M, Li X, Xu J, Jiang C, Zhao W, Ye F, Zhu W, Meng L, Lu S, Holmdahl R. Reactive oxygen species regulation by NCF1 governs ferroptosis susceptibility of Kupffer cells to MASH. Cell Metab 2024; 36:1745-1763.e6. [PMID: 38851189 DOI: 10.1016/j.cmet.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 11/17/2023] [Accepted: 05/13/2024] [Indexed: 06/10/2024]
Abstract
Impaired self-renewal of Kupffer cells (KCs) leads to inflammation in metabolic dysfunction-associated steatohepatitis (MASH). Here, we identify neutrophil cytosolic factor 1 (NCF1) as a critical regulator of iron homeostasis in KCs. NCF1 is upregulated in liver macrophages and dendritic cells in humans with metabolic dysfunction-associated steatotic liver disease and in MASH mice. Macrophage NCF1, but not dendritic cell NCF1, triggers KC iron overload, ferroptosis, and monocyte-derived macrophage infiltration, thus aggravating MASH progression. Mechanistically, elevated oxidized phospholipids induced by macrophage NCF1 promote Toll-like receptor (TLR4)-dependent hepatocyte hepcidin production, leading to increased KC iron deposition and subsequent KC ferroptosis. Importantly, the human low-functional polymorphic variant NCF190H alleviates KC ferroptosis and MASH in mice. In conclusion, macrophage NCF1 impairs iron homeostasis in KCs by oxidizing phospholipids, triggering hepatocyte hepcidin release and KC ferroptosis in MASH, highlighting NCF1 as a therapeutic target for improving KC fate and limiting MASH progression.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Infectious Diseases and National-Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China; Institute of Molecular and Translational Medicine and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Yu Wang
- Institute of Molecular and Translational Medicine and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Meiyang Fan
- Institute of Molecular and Translational Medicine and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Yanglong Guan
- Institute of Molecular and Translational Medicine and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Wentao Zhang
- Institute of Molecular and Translational Medicine and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Fumeng Huang
- Institute of Molecular and Translational Medicine and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Zhengqiang Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Xiaomeng Li
- Institute of Molecular and Translational Medicine and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Bingyu Yuan
- Institute of Molecular and Translational Medicine and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Wenbin Liu
- Institute of Molecular and Translational Medicine and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Manman Geng
- Department of Infectious Diseases and National-Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Xiaowei Li
- Department of Infectious Diseases and National-Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Jing Xu
- Institute of Molecular and Translational Medicine and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Congshan Jiang
- Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Xi'an 710003, Shaanxi, China
| | - Wenjuan Zhao
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Feng Ye
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Wenhua Zhu
- Institute of Molecular and Translational Medicine and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China.
| | - Liesu Meng
- Department of Infectious Diseases and National-Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China; Institute of Molecular and Translational Medicine and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, China.
| | - Shemin Lu
- Institute of Molecular and Translational Medicine and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, China
| | - Rikard Holmdahl
- Department of Infectious Diseases and National-Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China; Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, China; Medical Inflammation Research Group, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
2
|
Venn-Watson S. The Cellular Stability Hypothesis: Evidence of Ferroptosis and Accelerated Aging-Associated Diseases as Newly Identified Nutritional Pentadecanoic Acid (C15:0) Deficiency Syndrome. Metabolites 2024; 14:355. [PMID: 39057678 PMCID: PMC11279173 DOI: 10.3390/metabo14070355] [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: 05/23/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Ferroptosis is a newly discovered form of cell death caused by the peroxidation of fragile fatty acids in cell membranes, which combines with iron to increase reactive oxygen species and disable mitochondria. Ferroptosis has been linked to aging-related conditions, including type 2 diabetes, cardiovascular disease, and nonalcoholic fatty liver disease (NAFLD). Pentadecanoic acid (C15:0), an odd-chain saturated fat, is an essential fatty acid with the primary roles of stabilizing cell membranes and repairing mitochondrial function. By doing so, C15:0 reverses the underpinnings of ferroptosis. Under the proposed "Cellular Stability Hypothesis", evidence is provided to show that cell membranes optimally need >0.4% to 0.64% C15:0 to support long-term health and longevity. A pathophysiology of a newly identified nutritional C15:0 deficiency syndrome ("Cellular Fragility Syndrome") is provided that demonstrates how C15:0 deficiencies (≤0.2% total circulating fatty acids) can increase susceptibilities to ferroptosis, dysmetabolic iron overload syndrome, type 2 diabetes, cardiovascular disease, and NAFLD. Further, evidence is provided that C15:0 supplementation can reverse the described C15:0 deficiency syndrome, including the key components of ferroptosis. Given the declining dietary intake of C15:0, especially among younger generations, there is a need for extensive studies to understand the potential breadth of Cellular Fragility Syndrome across populations.
Collapse
Affiliation(s)
- Stephanie Venn-Watson
- Seraphina Therapeutics Inc., San Diego, CA 92106, USA;
- Epitracker Inc., San Diego, CA 92106, USA
| |
Collapse
|
3
|
He Q, Zheng Q, Diao H, Li M, Zhu Q, Fang F, Cui W. The role of body mass index on the association between the energy-adjusted dietary inflammatory index and hyperuricemia: a mediation analysis based on NHANES (2007-2016). Int J Obes (Lond) 2024; 48:339-345. [PMID: 37989765 DOI: 10.1038/s41366-023-01418-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/15/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND The Energy-Adjusted Dietary Inflammatory Index (E-DII) is related to both body mass index (BMI) and hyperuricemia. However, the association among BMI, hyperuricemia and DII is yet to be fully elucidated. The purpose of this study is to explore the role of BMI in the relationship between E-DII and hyperuricemia in the American population. METHODS A cross-sectional study was conducted using data from the National Health and Nutrition Examination Survey (NHANES) spanning from 2007 to 2016, with a sample size of 10,571 participants. The study used a weighted logistic regression model and a generalized additive model (GAM) to explore the associations among BMI, hyperuricemia and E-DII. Furthermore, mediation analysis was utilized to illustrate the mediating relationships among these variables. RESULTS The results of the study indicated that a higher E-DII was related to an increased risk of hyperuricemia. The association between E-DII and hyperuricemia was partially mediated by BMI. CONCLUSIONS E-DII is associated with hyperuricemia. BMI mediates the relationship between E-DII and hyperuricemia among Americans, which provides crucial information for the prevention of hyperuricemia.
Collapse
Affiliation(s)
- Qingzhen He
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, P. R. China
| | - Qingzhao Zheng
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, P. R. China
| | - Houze Diao
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, P. R. China
| | - Mingyuan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, P. R. China
| | - Qing Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, P. R. China
| | - Fang Fang
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, P. R. China.
| | - Weiwei Cui
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, P. R. China.
| |
Collapse
|
4
|
Zhang H, Sumbria RK, Chang R, Sun J, Cribbs DH, Holmes TC, Fisher MJ, Xu X. Erythrocyte-brain endothelial interactions induce microglial responses and cerebral microhemorrhages in vivo. J Neuroinflammation 2023; 20:265. [PMID: 37968737 PMCID: PMC10647121 DOI: 10.1186/s12974-023-02932-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/13/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Cerebral microhemorrhages (CMH) are associated with stroke, cognitive decline, and normal aging. Our previous study shows that the interaction between oxidatively stressed red blood cells (RBC) and cerebral endothelium may underlie CMH development. However, the real-time examination of altered RBC-brain endothelial interactions in vivo, and their relationship with clearance of stalled RBC, microglial responses, and CMH development, has not been reported. METHODS RBC were oxidatively stressed using tert-butylhydroperoxide (t-BHP), fluorescently labeled and injected into adult Tie2-GFP mice. In vivo two-photon imaging and ex vivo confocal microscopy were used to evaluate the temporal profile of RBC-brain endothelial interactions associated with oxidatively stressed RBC. Their relationship with microglial activation and CMH was examined with post-mortem histology. RESULTS Oxidatively stressed RBC stall significantly and rapidly in cerebral vessels in mice, accompanied by decreased blood flow velocity which recovers at 5 days. Post-mortem histology confirms significantly greater RBC-cerebral endothelial interactions and microglial activation at 24 h after t-BHP-treated RBC injection, which persist at 7 days. Furthermore, significant CMH develop in the absence of blood-brain barrier leakage after t-BHP-RBC injection. CONCLUSIONS Our in vivo and ex vivo findings show the stalling and clearance of oxidatively stressed RBC in cerebral capillaries, highlighting the significance of microglial responses and altered RBC-brain endothelial interactions in CMH development. Our study provides novel mechanistic insight into CMH associated with pathological conditions with increased RBC-brain endothelial interactions.
Collapse
Affiliation(s)
- Hai Zhang
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Rachita K Sumbria
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA.
- Department of Neurology, University of California, Irvine, CA, 92697, USA.
| | - Rudy Chang
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - Jiahong Sun
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - David H Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Todd C Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
| | - Mark J Fisher
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA.
- Department of Neurology, University of California, Irvine, CA, 92697, USA.
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA.
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA.
- Beckman Laser Institute, University of California, Irvine, CA, 92697, USA.
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA, 92697, USA.
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA.
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA.
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA.
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
5
|
Park JB, Ko K, Baek YH, Kwon WY, Suh S, Han SH, Kim YH, Kim HY, Yoo YH. Pharmacological Prevention of Ectopic Erythrophagocytosis by Cilostazol Mitigates Ferroptosis in NASH. Int J Mol Sci 2023; 24:12862. [PMID: 37629045 PMCID: PMC10454295 DOI: 10.3390/ijms241612862] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/04/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Hepatic iron overload (HIO) is a hallmark of nonalcoholic fatty liver disease (NAFLD) with a poor prognosis. Recently, the role of hepatic erythrophagocytosis in NAFLD is emerging as a cause of HIO. We undertook various assays using human NAFLD patient pathology samples and an in vivo nonalcoholic steatohepatitis (NASH) mouse model named STAMTM. To make the in vitro conditions comparable to those of the in vivo NASH model, red blood cells (RBCs) and platelets were suspended and subjected to metabolic and inflammatory stresses. An insert-coculture system, in which activated THP-1 cells and RBCs are separated from HepG2 cells by a porous membrane, was also employed. Through various analyses in this study, the effect of cilostazol was examined. The NAFLD activity score, including steatosis, ballooning degeneration, inflammation, and fibrosis, was increased in STAMTM mice. Importantly, hemolysis occurred in the serum of STAMTM mice. Although cilostazol did not improve lipid or glucose profiles, it ameliorated hepatic steatosis and inflammation in STAMTM mice. Platelets (PLTs) played an important role in increasing erythrophagocytosis in the NASH liver. Upregulated erythrophagocytosis drives cells into ferroptosis, resulting in liver cell death. Cilostazol inhibited the augmentation of PLT and RBC accumulation. Cilostazol prevented the PLT-induced increase in ectopic erythrophagocytosis in in vivo and in vitro NASH models. Cilostazol attenuated ferroptosis of hepatocytes and phagocytosis of RBCs by THP-1 cells. Augmentation of hepatic erythrophagocytosis by activated platelets in NASH exacerbates HIO. Cilostazol prevents ectopic erythrophagocytosis, mitigating HIO-mediated ferroptosis in NASH models.
Collapse
Affiliation(s)
- Joon Beom Park
- Department of Anatomy and Cell Biology, Dong-A University College of Medicine, Busan 49201, Republic of Korea; (J.B.P.); (K.K.); (W.Y.K.)
| | - Kangeun Ko
- Department of Anatomy and Cell Biology, Dong-A University College of Medicine, Busan 49201, Republic of Korea; (J.B.P.); (K.K.); (W.Y.K.)
| | - Yang Hyun Baek
- Department of Gastroenterology, Dong-A University College of Medicine, Busan 49201, Republic of Korea;
| | - Woo Young Kwon
- Department of Anatomy and Cell Biology, Dong-A University College of Medicine, Busan 49201, Republic of Korea; (J.B.P.); (K.K.); (W.Y.K.)
| | - Sunghwan Suh
- Department of Endocrinology, Dong-A University College of Medicine, Busan 49201, Republic of Korea;
| | - Song-Hee Han
- Department of Pathology, Dong-A University College of Medicine, Busan 49201, Republic of Korea;
| | - Yun Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Hye Young Kim
- Department of Anatomy, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Young Hyun Yoo
- Department of Anatomy and Cell Biology, Dong-A University College of Medicine, Busan 49201, Republic of Korea; (J.B.P.); (K.K.); (W.Y.K.)
| |
Collapse
|
6
|
Kouroumalis E, Tsomidis I, Voumvouraki A. Iron as a therapeutic target in chronic liver disease. World J Gastroenterol 2023; 29:616-655. [PMID: 36742167 PMCID: PMC9896614 DOI: 10.3748/wjg.v29.i4.616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/03/2022] [Accepted: 12/31/2022] [Indexed: 01/20/2023] Open
Abstract
It was clearly realized more than 50 years ago that iron deposition in the liver may be a critical factor in the development and progression of liver disease. The recent clarification of ferroptosis as a specific form of regulated hepatocyte death different from apoptosis and the description of ferritinophagy as a specific variation of autophagy prompted detailed investigations on the association of iron and the liver. In this review, we will present a brief discussion of iron absorption and handling by the liver with emphasis on the role of liver macrophages and the significance of the iron regulators hepcidin, transferrin, and ferritin in iron homeostasis. The regulation of ferroptosis by endogenous and exogenous mod-ulators will be examined. Furthermore, the involvement of iron and ferroptosis in various liver diseases including alcoholic and non-alcoholic liver disease, chronic hepatitis B and C, liver fibrosis, and hepatocellular carcinoma (HCC) will be analyzed. Finally, experimental and clinical results following interventions to reduce iron deposition and the promising manipulation of ferroptosis will be presented. Most liver diseases will be benefited by ferroptosis inhibition using exogenous inhibitors with the notable exception of HCC, where induction of ferroptosis is the desired effect. Current evidence mostly stems from in vitro and in vivo experimental studies and the need for well-designed future clinical trials is warranted.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Liver Research Laboratory, University of Crete Medical School, Heraklion 71003, Greece
| | - Ioannis Tsomidis
- First Department of Internal Medicine, AHEPA University Hospital, Thessaloniki 54621, Greece
| | - Argyro Voumvouraki
- First Department of Internal Medicine, AHEPA University Hospital, Thessaloniki 54621, Greece
| |
Collapse
|
7
|
Deng Q, Wang W, Zhang L, Chen L, Zhang Q, Zhang Y, He S, Li J. Gougunao tea polysaccharides ameliorate high-fat diet-induced hyperlipidemia and modulate gut microbiota. Food Funct 2023; 14:703-719. [PMID: 36511170 DOI: 10.1039/d2fo01828d] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Many natural polysaccharides have been proven to have ameliorative effects on high-fat diet-induced hyperlipidemia with fewer side effects. However, similar data on Gougunao tea polysaccharides remain obscure. In this study, we aimed to investigate the role of Gougunao tea polysaccharides (GTP40) in the alleviation of hyperlipidemia and regulation of gut microbiota in C57BL/6J mice induced by a high-fat diet. The results indicated that GTP40 intervention inhibited the abnormal growth of body weight and the excessive accumulation of lipid droplets in the livers and ameliorated the biochemical parameters of serum/liver related to lipid metabolism in hyperlipidemia mice. The elevated levels of antioxidant enzyme and anti-inflammation cytokine in serum, as well as the up-regulating anti-inflammation gene in the liver, reflected that GTP40 might mitigate the oxidative and inflammatory stress induced by a high-fat diet. In addition, GTP40 could modulate the composition, abundance, and diversity of gut microbiota in hyperlipidemia mice. Besides, Spearman's correlation analysis implied that GTP40 intervention could enrich beneficial bacteria (e.g., Akkermansia, Bacteroides, Roseburia, and Alistipes), and decrease harmful bacteria (e.g., Blautia, Faecalibaculum, Streptococcus, and norank_f_Desulfovibrionaceae), which were correlated with the lipid metabolic parameters associated with hyperlipidemia. Moreover, it also indicated that there was a significant correlation between gut microbiota and SCFAs. Thus, GTP40 may be a novel strategy against fat accumulation, oxidative stress, and inflammation, as well as restoring the normal microbial balance of the gut in hyperlipidemia mice.
Collapse
Affiliation(s)
- Qihuan Deng
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Wenjun Wang
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Lieyuan Zhang
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China. .,Technical Center of Nanchang Customs, Nanchang 330038, China
| | - Lingli Chen
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Qingfeng Zhang
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Ying Zhang
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Sichen He
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Jingen Li
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| |
Collapse
|
8
|
Papadopoulos C, Anagnostopoulos K, Tsiptsios D, Karatzetzou S, Liaptsi E, Lazaridou IZ, Kokkotis C, Makri E, Ioannidou M, Aggelousis N, Vadikolias K. Unexplored Roles of Erythrocytes in Atherothrombotic Stroke. Neurol Int 2023; 15:124-139. [PMID: 36810466 PMCID: PMC9944955 DOI: 10.3390/neurolint15010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Stroke constitutes the second highest cause of morbidity and mortality worldwide while also impacting the world economy, triggering substantial financial burden in national health systems. High levels of blood glucose, homocysteine, and cholesterol are causative factors for atherothrombosis. These molecules induce erythrocyte dysfunction, which can culminate in atherosclerosis, thrombosis, thrombus stabilization, and post-stroke hypoxia. Glucose, toxic lipids, and homocysteine result in erythrocyte oxidative stress. This leads to phosphatidylserine exposure, promoting phagocytosis. Phagocytosis by endothelial cells, intraplaque macrophages, and vascular smooth muscle cells contribute to the expansion of the atherosclerotic plaque. In addition, oxidative stress-induced erythrocytes and endothelial cell arginase upregulation limit the pool for nitric oxide synthesis, leading to endothelial activation. Increased arginase activity may also lead to the formation of polyamines, which limit the deformability of red blood cells, hence facilitating erythrophagocytosis. Erythrocytes can also participate in the activation of platelets through the release of ADP and ATP and the activation of death receptors and pro-thrombin. Damaged erythrocytes can also associate with neutrophil extracellular traps and subsequently activate T lymphocytes. In addition, reduced levels of CD47 protein in the surface of red blood cells can also lead to erythrophagocytosis and a reduced association with fibrinogen. In the ischemic tissue, impaired erythrocyte 2,3 biphosphoglycerate, because of obesity or aging, can also favor hypoxic brain inflammation, while the release of damage molecules can lead to further erythrocyte dysfunction and death.
Collapse
Affiliation(s)
- Charalampos Papadopoulos
- Laboratory of Biochemistry, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Konstantinos Anagnostopoulos
- Laboratory of Biochemistry, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Dimitrios Tsiptsios
- Department of Neurology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
- Correspondence:
| | - Stella Karatzetzou
- Department of Neurology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Eirini Liaptsi
- Department of Neurology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | | | - Christos Kokkotis
- Department of Physical Education and Sport Science, Democritus University of Thrace, 69100 Komotini, Greece
| | - Evangelia Makri
- Department of Physical Education and Sport Science, Democritus University of Thrace, 69100 Komotini, Greece
| | - Maria Ioannidou
- Department of Physical Education and Sport Science, Democritus University of Thrace, 69100 Komotini, Greece
| | - Nikolaos Aggelousis
- Department of Physical Education and Sport Science, Democritus University of Thrace, 69100 Komotini, Greece
| | | |
Collapse
|
9
|
Lei T, Zhang J, Zhang Q, Ma X, Xu Y, Zhao Y, Zhang L, Lu Z, Zhao Y. Defining newly formed and tissue-resident bone marrow-derived macrophages in adult mice based on lysozyme expression. Cell Mol Immunol 2022; 19:1333-1346. [PMID: 36348079 PMCID: PMC9708686 DOI: 10.1038/s41423-022-00936-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 10/08/2022] [Indexed: 11/11/2022] Open
Abstract
Tissue-resident macrophages are derived from different precursor cells and display different phenotypes. Reconstitution of the tissue-resident macrophages of inflamed or damaged tissues in adults can be achieved by bone marrow-derived monocytes/macrophages. Using lysozyme (Lysm)-GFP-reporter mice, we found that alveolar macrophages (AMs), Kupffer cells, red pulp macrophages (RpMacs), and kidney-resident macrophages were Lysm-GFP-, whereas all monocytes in the fetal liver, adult bone marrow, and blood were Lysm-GFP+. Donor-derived Lysm-GFP+ resident macrophages gradually became Lysm-GFP- in recipients and developed gene expression profiles characteristic of tissue-resident macrophages. Thus, Lysm may be used to distinguish newly formed and long-term surviving tissue-resident macrophages that were derived from bone marrow precursor cells in adult mice under pathological conditions. Furthermore, we found that Irf4 might be essential for resident macrophage differentiation in all tissues, while cytokine and receptor pathways, mTOR signaling pathways, and fatty acid metabolic processes predominantly regulated the differentiation of RpMacs, Kupffer cells, and kidney macrophages, respectively. Deficiencies in ST2, mechanistic target of rapamycin (mTOR) and fatty acid-binding protein 5 (FABP5) differentially impaired the differentiation of tissue-resident macrophages from bone marrow-derived monocytes/macrophages in the lungs, liver, and kidneys. These results indicate that a combination of shared and unique signaling pathways coordinately shape tissue-resident macrophage differentiation in various tissues.
Collapse
Affiliation(s)
- Tong Lei
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jiayu Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Cunji Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Qian Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Cunji Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Xinran Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yanan Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yang Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lianfeng Zhang
- Key Laboratory of Human Diseases Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China.
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Cunji Medical School, University of Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
| |
Collapse
|
10
|
Hino K, Yanatori I, Hara Y, Nishina S. Iron and liver cancer: an inseparable connection. FEBS J 2022; 289:7810-7829. [PMID: 34543507 DOI: 10.1111/febs.16208] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/17/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023]
Abstract
Iron is an essential element for all organisms. Iron-containing proteins play critical roles in cellular functions. The biological importance of iron is largely attributable to its chemical properties as a transitional metal. However, an excess of 'free' reactive iron damages the macromolecular components of cells and cellular DNA through the production of harmful free radicals. On the contrary, most of the body's excess iron is stored in the liver. Not only hereditary haemochromatosis but also some liver diseases with mild-to-moderate hepatic iron accumulation, such as chronic hepatitis C, alcoholic liver disease and nonalcoholic steatohepatitis, are associated with a high risk for liver cancer development. These findings have attracted attention to the causative and promotive roles of iron in the development of liver cancer. In the last decade, accumulating evidence regarding molecules regulating iron metabolism or iron-related cell death programmes such as ferroptosis has shed light on the relationship between hepatic iron accumulation and hepatocarcinogenesis. In this review, we briefly present the current molecular understanding of iron regulation in the liver. Next, we describe the mechanisms underlying dysregulated iron metabolism depending on the aetiology of liver diseases. Finally, we discuss the causative and promotive roles of iron in cancer development.
Collapse
Affiliation(s)
- Keisuke Hino
- Department of Hepatology and Pancreatology, Kawasaki Medical School, Kurashiki, Japan
| | - Izumi Yanatori
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Japan
| | - Yuichi Hara
- Department of Hepatology and Pancreatology, Kawasaki Medical School, Kurashiki, Japan
| | - Sohji Nishina
- Department of Hepatology and Pancreatology, Kawasaki Medical School, Kurashiki, Japan
| |
Collapse
|
11
|
Ma C, Han L, Zhu Z, Heng Pang C, Pan G. Mineral metabolism and ferroptosis in non-alcoholic fatty liver diseases. Biochem Pharmacol 2022; 205:115242. [PMID: 36084708 DOI: 10.1016/j.bcp.2022.115242] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/02/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become the most prevalent chronic liver disease worldwide. Minerals including iron, copper, zinc, and selenium, fulfil an essential role in various biochemical processes. Moreover, the identification of ferroptosis and cuproptosis further underscores the importance of intracellular mineral homeostasis. However, perturbation of minerals has been frequently reported in patients with NAFLD and related diseases. Interestingly, studies have attempted to establish an association between mineral disorders and NAFLD pathological features, including oxidative stress, mitochondrial dysfunction, inflammatory response, and fibrogenesis. In this review, we aim to provide an overview of the current understanding of mineral metabolism (i.e., absorption, utilization, and transport) and mineral interactions in the pathogenesis of NAFLD. More importantly, this review highlights potential therapeutic strategies, challenges, future directions for targeting mineral metabolism in the treatment of NAFLD.
Collapse
Affiliation(s)
- Chenhui Ma
- Department of Chemical and Environmental Engineering, University of Nottingham Ningbo China, Ningbo 315100, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Han
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zheying Zhu
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, UK.
| | - Cheng Heng Pang
- Department of Chemical and Environmental Engineering, University of Nottingham Ningbo China, Ningbo 315100, China.
| | - Guoyu Pan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
12
|
Šrajer Gajdošik M, Kovač Peić A, Begić M, Grbčić P, Brilliant KE, Hixson DC, Josić D. Possible Role of Extracellular Vesicles in Hepatotoxicity of Acetaminophen. Int J Mol Sci 2022; 23:8870. [PMID: 36012131 PMCID: PMC9408656 DOI: 10.3390/ijms23168870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 07/29/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022] Open
Abstract
We examined proteomic profiles of rat liver extracellular vesicles (EVs) shed following treatment with a sub-toxic dose (500 mg/kg) of the pain reliever drug, acetaminophen (APAP). EVs representing the entire complement of hepatic cells were isolated after perfusion of the intact liver and analyzed with LC-MS/MS. The investigation was focused on revealing the function and cellular origin of identified EVs proteins shed by different parenchymal and non-parenchymal liver cells and their possible role in an early response of this organ to a toxic environment. Comparison of EV proteomic profiles from control and APAP-treated animals revealed significant differences. Alpha-1-macroglobulin and members of the cytochrome P450 superfamily were highly abundant proteins in EVs shed by the normal liver. In contrast, proteins like aminopeptidase N, metalloreductase STEAP4, different surface antigens like CD14 and CD45, and most members of the annexin family were detected only in EVs that were shed by livers of APAP-treated animals. In EVs from treated livers, there was almost a complete disappearance of members of the cytochrome P450 superfamily and a major decrease in other enzymes involved in the detoxification of xenobiotics. Additionally, there were proteins that predominated in non-parenchymal liver cells and in the extracellular matrix, like fibronectin, receptor-type tyrosine-protein phosphatase C, and endothelial type gp91. These differences indicate that even treatment with a sub-toxic concentration of APAP initiates dramatic perturbation in the function of this vital organ.
Collapse
Affiliation(s)
| | | | - Marija Begić
- Faculty of Medicine, University Juraj Dobrila of Pula, 52100 Pula, Croatia
| | - Petra Grbčić
- Faculty of Medicine, University Juraj Dobrila of Pula, 52100 Pula, Croatia
| | - Kate E. Brilliant
- Proteomics Core, COBRE CCRD, Rhode Island Hospital, Providence, RI 02903, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Douglas C. Hixson
- Proteomics Core, COBRE CCRD, Rhode Island Hospital, Providence, RI 02903, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Djuro Josić
- Faculty of Medicine, University Juraj Dobrila of Pula, 52100 Pula, Croatia
- Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| |
Collapse
|
13
|
Papadopoulos C, Spourita E, Mimidis K, Kolios G, Tentes L, Anagnostopoulos K. Nonalcoholic Fatty Liver Disease Patients Exhibit Reduced CD47 and Increased Sphingosine, Cholesterol, and Monocyte Chemoattractant Protein-1 Levels in the Erythrocyte Membranes. Metab Syndr Relat Disord 2022; 20:377-383. [PMID: 35532955 DOI: 10.1089/met.2022.0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: Nonalcoholic fatty liver disease (NAFLD) constitutes a significant cause of deaths, liver transplantations, and economic costs worldwide. Despite extended research, investigations on the role of erythrocytes are scarce. Red blood cells from experimental animals and human patients with NAFLD present phosphatidylserine exposure, which is then recognized by Kupffer cells. This event leads to erythrophagocytosis and amplification of inflammation through iron disposition. In addition, it has been shown that erythrocytes from NAFLD patients release the chemokine monocyte chemoattractant protein-1 (MCP1), leading to increased tumor necrosis factor alpha release from macrophages RAW 264.7. However, erythrophagocytosis can also be caused by reduced CD47 levels. Moreover, increased MCP1 release could be either signal-induced or caused by higher MCP1 levels on the erythrocyte membrane. Finally, erythrocyte efferocytosis could provide additional inflammatory metabolites. Methods: In this study, we measured the erythrocyte membrane levels of CD47 and MCP1 by enzyme-linked immunosorbent assay, and cholesterol and sphingosine with thin-layer chromatography. Eighteen patients (8 men and 10 women, aged 56.7 ± 11.5 years) and 14 healthy controls (7 men and 7 women, aged 39.3 ± 15.6 years) participated in our study. Results: The erythrocyte CD47 levels were decreased in the erythrocyte membranes of NAFLD patients (844 ± 409 pg/mL) compared with healthy controls (2969 ± 1936 pg/mL) with P = 0.012. Levels of MCP1 increased in NAFLD patients (389 ± 255 pg/mL) compared with healthy controls (230 ± 117 pg/mL) with P = 0.0274, but low statistical power. Moreover, in erythrocyte membranes, there was a statistically significant accumulation of sphingosine and cholesterol in NAFLD patients compared with healthy controls. Conclusions: Our results imply that erythrocytes release chemotactic "find me" signals (MCP1) while containing reduced "do not eat me" signals (CD47). These molecules can lead to erythrophagocytosis. Next, increased "goodbye" signals (sphingosine and cholesterol) could augment inflammation by metabolic reprogramming.
Collapse
Affiliation(s)
- Charalampos Papadopoulos
- Laboratory of Biochemistry, Department of Basic Sciences, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Eleftheria Spourita
- Laboratory of Biochemistry, Department of Basic Sciences, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Konstantinos Mimidis
- First Department of Internal Medicine, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - George Kolios
- Laboratory of Pharmacology, Department of Basic Sciences, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Loannis Tentes
- Laboratory of Biochemistry, Department of Basic Sciences, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Konstantinos Anagnostopoulos
- Laboratory of Biochemistry, Department of Basic Sciences, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
14
|
Red Blood Cell-Conditioned Media from Non-Alcoholic Fatty Liver Disease Patients Contain Increased MCP1 and Induce TNF-α Release. Rep Biochem Mol Biol 2022; 11:54-62. [PMID: 35765536 PMCID: PMC9208556 DOI: 10.52547/rbmb.11.1.54] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/12/2021] [Indexed: 01/11/2023]
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) constitutes a global pandemic. An intricate network among cytokines and lipids possesses a central role in NAFLD pathogenesis. Red blood cells comprise an important source of both cytokines and signaling lipids and have an important role in molecular crosstalk during immunometabolic deregulation. However, their role in NAFLD has not been thoroughly investigated. Methods Conditioned media from erythrocytes derived from 10 NAFLD patients (4 men, 6 women, aged 57.875±15.16) and 10 healthy controls (4 men, 6 women, aged 39.3±15.55) was analyzed for the cytokines IFN-γ, TNF-α, CCL2, CCL5, IL-8, IL-1β, IL-12p40, IL-17, MIP-1β, the signaling lipids sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA), and cholesterol. Their effect on the cytokine profile released by RAW 264.7 macrophages was also studied. Results MCP1 levels were greater in conditioned growth medium from NAFLD patient erythrocytes than in that from healthy controls (37±40 vs 6.51±5.63 pg/ml). No statistically significant differences were found between patients and healthy controls with regard to S1P, LPA, cholesterol, or eight other cytokines. TNF-a release by RAW 264.7 cells was greater after incubation with patient-derived erythrocyte-conditioned medium than in medium without RAW 264.7 cells from either healthy or NAFLD subjects. Conclusion Erythrocytes may contribute to liver infiltration by monocytes, and macrophage activation, partially due to CCL2 release, in the context of NAFLD..
Collapse
|
15
|
Li LX, Guo FF, Liu H, Zeng T. Iron overload in alcoholic liver disease: underlying mechanisms, detrimental effects, and potential therapeutic targets. Cell Mol Life Sci 2022; 79:201. [PMID: 35325321 PMCID: PMC11071846 DOI: 10.1007/s00018-022-04239-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023]
Abstract
Alcoholic liver disease (ALD) is a global public health challenge due to the high incidence and lack of effective therapeutics. Evidence from animal studies and ALD patients has demonstrated that iron overload is a hallmark of ALD. Ethanol exposure can promote iron absorption by downregulating the hepcidin expression, which is probably mediated by inducing oxidative stress and promoting erythropoietin (EPO) production. In addition, ethanol may enhance iron uptake in hepatocytes by upregulating the expression of transferrin receptor (TfR). Iron overload in the liver can aggravate ethanol-elicited liver damage by potentiating oxidative stress via Fenton reaction, promoting activation of Kupffer cells (KCs) and hepatic stellate cells (HSCs), and inducing a recently discovered programmed iron-dependent cell death, ferroptosis. This article reviews the current knowledge of iron metabolism, regulators of iron homeostasis, the mechanism of ethanol-induced iron overload, detrimental effects of iron overload in the liver, and potential therapeutic targets.
Collapse
Affiliation(s)
- Long-Xia Li
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Fang-Fang Guo
- Department of Pharmacy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Hong Liu
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
16
|
Validation and application of a protocol for the extraction and quantitative analysis of sphingomyelin in erythrocyte membranes of patients with non-alcoholic fatty liver disease. JPC-J PLANAR CHROMAT 2021. [DOI: 10.1007/s00764-021-00127-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
17
|
Imajo K, Kessoku T, Honda Y, Hasegawa S, Tomeno W, Ogawa Y, Motosugi U, Saigusa Y, Yoneda M, Kirikoshi H, Yamanaka S, Utsunomiya D, Saito S, Nakajima A. MRI-Based Quantitative R2 * Mapping at 3 Tesla Reflects Hepatic Iron Overload and Pathogenesis in Nonalcoholic Fatty Liver Disease Patients. J Magn Reson Imaging 2021; 55:111-125. [PMID: 34184822 DOI: 10.1002/jmri.27810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The role of hepatic iron overload (HIO) in nonalcoholic fatty liver disease (NAFLD) pathogenesis has not been fully elucidated. PURPOSE This study aimed to investigate the effect of HIO and examine the diagnostic usefulness of magnetic resonance imaging (MRI)-based R2* quantification in evaluating hepatic iron content (HIC) and pathological findings in NAFLD. STUDY TYPE Prospective and retrospective. POPULATION A prospective study of 168 patients (age, 57.2 ± 15.0; male/female, 80/88) and a retrospective validation study of 202 patients (age, 57.0 ± 14.4; male/female, 113/89) with liver-biopsy-confirmed NAFLD were performed. FIELD STRENGTH/SEQUENCE 3 T; chemical-shift encoded multi-echo gradient echo. ASSESSMENT Using liver tissues obtained by liver biopsy, HIC was prospectively evaluated in 168 patients by atomic absorption spectrometry. Diagnostic accuracies of HIC and R2* for grading hepatic inflammation plus ballooning (HIB) as an indicator of NAFLD activity were assessed. STATISTICAL TESTS Student's t-test and analysis of variance (ANOVA) with Scheffe's multiple testing correction for univariate comparisons; multivariate logistic analysis. P-value less than 0.05 is statistically significant. RESULTS HIC was significantly correlated with HIB grades (r = 0.407). R2* was significantly correlated with HIC (r = 0.557) and HIB grades (r = 0.569). R2* mapped an area under the receiver operating characteristic (AUROC; 0.774) for HIC ≥808 ng/mL (median value) with cutoff value of 62.5 s-1 . In addition, R2* mapped AUROC of HIB for grades ≥3 was 0.799 with cutoff value of 58.5 s-1 . When R2* was <62.5 s-1 , R2* correlated weakly with HIC (r = 0.372) as it was affected by fat deposition and did not correlate with HIB grades (P = 0.052). Conversely, when R2* was ≥62.5 s-1 , a significant correlation of R2* with HIC (r = 0.556) and with HIB grades was observed (P < 0.0001) with being less affected by fat deposition. DATA CONCLUSION R2* ≥ 62.5 s-1 is a promising modality for non-invasive diagnosis of clinically important high grades (≥3) of HIB associated with increased HIC. LEVEL OF EVIDENCE 1 TECHNICAL EFFICACY STAGE: 2.
Collapse
Affiliation(s)
- Kento Imajo
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takaomi Kessoku
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yasushi Honda
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Sho Hasegawa
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Wataru Tomeno
- Department of Gastroenterology, International University of Health and Welfare Atami Hospital, Atami, Japan
| | - Yuji Ogawa
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Utaroh Motosugi
- Department of Radiology, University of Yamanashi, Chuo, Japan
| | - Yusuke Saigusa
- Department of Biostatistics, Yokohama City University School of Medicine, Yokohama, Japan
| | - Masato Yoneda
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroyuki Kirikoshi
- Department of Clinical Laboratory, Yokohama City University Hospital, Yokohama, Japan
| | - Shoji Yamanaka
- Anatomic and Clinical Pathology Department, Yokohama City University Hospital, Yokohama, Japan
| | - Daisuke Utsunomiya
- Department of Radiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoru Saito
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
18
|
Xia Y, Li Y, Wu X, Zhang Q, Chen S, Ma X, Yu M. Ironing Out the Details: How Iron Orchestrates Macrophage Polarization. Front Immunol 2021; 12:669566. [PMID: 34054839 PMCID: PMC8149954 DOI: 10.3389/fimmu.2021.669566] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Iron fine-tunes innate immune responses, including macrophage inflammation. In this review, we summarize the current understanding about the iron in dictating macrophage polarization. Mechanistically, iron orchestrates macrophage polarization through several aspects, including cellular signaling, cellular metabolism, and epigenetic regulation. Therefore, iron modulates the development and progression of multiple macrophage-associated diseases, such as cancer, atherosclerosis, and liver diseases. Collectively, this review highlights the crucial role of iron for macrophage polarization, and indicates the potential application of iron supplementation as an adjuvant therapy in different inflammatory disorders relative to the balance of macrophage polarization.
Collapse
Affiliation(s)
- Yaoyao Xia
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yikun Li
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaoyan Wu
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qingzhuo Zhang
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Siyuan Chen
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xianyong Ma
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Miao Yu
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
19
|
Papadopoulos C, Tentes I, Anagnostopoulos K. Red Blood Cell Dysfunction in Non-Alcoholic Fatty Liver Disease: Marker and Mediator of Molecular Mechanisms. MÆDICA 2021; 15:513-516. [PMID: 33603909 DOI: 10.26574/maedica.2020.15.4.513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Despite efforts to unravel the pathogenetic mechanisms of non-alcoholic fatty liver disease (NAFLD), there is still a need for approved treatments and biomarkers. Interestingly, red blood cells present alterations in their characteristics during NAFLD. The phosphatidylcholine to phosphatidylethanolamine ratio, fatty acid profile, red blood cell count and red cell distribution width reflect molecular changes that are taking place in the liver. In addition, glycosylated hemoglobin, chemokine binding and release, and phosphatidylserine exposure actively participate in NAFLD pathogenesis. In this review, we describe the neglected red blood cell dysfunction in NAFLD, with the aim to unveil potent biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Charalampos Papadopoulos
- Laboratory of Biochemistry, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Ioannis Tentes
- Laboratory of Biochemistry, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | | |
Collapse
|
20
|
Jorgačević B, Vučević D, Samardžić J, Mladenović D, Vesković M, Vukićević D, Ješić R, Radosavljević T. The Effect of CB1 Antagonism on Hepatic Oxidative/Nitrosative Stress and Inflammation in Nonalcoholic Fatty Liver Disease. Curr Med Chem 2021; 28:169-180. [PMID: 32124686 DOI: 10.2174/0929867327666200303122734] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/17/2019] [Accepted: 01/25/2020] [Indexed: 02/08/2023]
Abstract
Dysfunction of the endocannabinoid system (ES) has been identified in nonalcoholic fatty liver disease (NAFLD) and associated metabolic disorders. Cannabinoid receptor type 1 (CB1) expression is largely dependent on nutritional status. Thus, individuals suffering from NAFLD and metabolic syndrome (MS) have a significant increase in ES activity. Furthermore, oxidative/ nitrosative stress and inflammatory process modulation in the liver are highly influenced by the ES. Numerous experimental studies indicate that oxidative and nitrosative stress in the liver is associated with steatosis and portal inflammation during NAFLD. On the other hand, inflammation itself may also contribute to reactive oxygen species (ROS) production due to Kupffer cell activation and increased nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity. The pathways by which endocannabinoids and their lipid-related mediators modulate oxidative stress and lipid peroxidation represent a significant area of research that could yield novel pharmaceutical strategies for the treatment of NAFLD. Cumulative evidence suggested that the ES, particularly CB1 receptors, may also play a role in inflammation and disease progression toward steatohepatitis. Pharmacological inactivation of CB1 receptors in NAFLD exerts multiple beneficial effects, particularly due to the attenuation of hepatic oxidative/nitrosative stress parameters and significant reduction of proinflammatory cytokine production. However, further investigations regarding precise mechanisms by which CB1 blockade influences the reduction of hepatic oxidative/nitrosative stress and inflammation are required before moving toward the clinical phase of the investigation.
Collapse
Affiliation(s)
- Bojan Jorgačević
- Institute of Pathophysiology ''Ljubodrag Buba Mihailović'', Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Danijela Vučević
- Institute of Pathophysiology ''Ljubodrag Buba Mihailović'', Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Janko Samardžić
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Dušan Mladenović
- Institute of Pathophysiology ''Ljubodrag Buba Mihailović'', Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Milena Vesković
- Institute of Pathophysiology ''Ljubodrag Buba Mihailović'', Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Dušan Vukićević
- Institute of Pathophysiology ''Ljubodrag Buba Mihailović'', Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Rada Ješić
- Institute of Digestive Diseases, Clinical Centre of Serbia, 11000 Belgrade, Serbia
| | - Tatjana Radosavljević
- Institute of Pathophysiology ''Ljubodrag Buba Mihailović'', Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
21
|
Saeed NM, Mansour AM, Allam S. Lycopene induces insulin signaling and alleviates fibrosis in experimental model of non-alcoholic fatty liver disease in rats. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
22
|
Papadopoulos C, Panopoulou M, Anagnostopoulos K, Tentes I. Immune and Metabolic Interactions of Human Erythrocytes: A Molecular Perspective. Endocr Metab Immune Disord Drug Targets 2020; 21:843-853. [PMID: 33148159 DOI: 10.2174/1871530320666201104115016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/15/2020] [Accepted: 09/24/2020] [Indexed: 11/22/2022]
Abstract
Apart from their main function as oxygen carriers in vertebrates, erythrocytes are also involved in immune regulation. By circulating throughout the body, the erythrocytes are exposed and interact with tissues that are damaged as a result of a disease. In this study, we summarize the literature regarding the contribution of erythrocytes to immune regulation and metabolism. Under the circumstances of a disease state, the erythrocytes may lose their antioxidant capacity and release Damage Associated Molecular Patterns, resulting in the regulation of innate and adaptive immunity. In addition, the erythrocytes scavenge and affect the levels of chemokines, circulating cell-free mtDNA, and C3b attached immune complexes. Furthermore, through surface molecules, erythrocytes control the function of T lymphocytes, macrophages, and dendritic cells. Through an array of enzymes, red blood cells contribute to the pool of blood's bioactive lipids. Finally, the erythrocytes contribute to reverse cholesterol transport through various mechanisms. Our study is highlighting overlooked molecular interactions between erythrocytes and immunity and metabolism, which could lead to the discovery of potent therapeutic targets for immunometabolic diseases.
Collapse
Affiliation(s)
| | - Maria Panopoulou
- Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | | | - Ioannis Tentes
- Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
23
|
Xu W, Xu YN, Zhang X, Xu Y, Jian X, Chen JM, Chen GF, Zhang H, Liu P, Mu YP. Hepatic stem cell Numb gene is a potential target of Huang Qi Decoction against cholestatic liver fibrosis. Sci Rep 2020; 10:17486. [PMID: 33060633 PMCID: PMC7566460 DOI: 10.1038/s41598-020-74324-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/30/2020] [Indexed: 12/13/2022] Open
Abstract
Numb is a negative regulator of Notch signal pathway. Previous study has demonstrated that Notch signal pathway activation is required for hepatic progenitor cell (HPC) differentiating into cholangiocytes in cholestatic liver fibrosis (CLF), and Huang Qi Decoction (HQD) could prevent CLF through inhibition of the Notch signal pathway. However, the role of Numb in HQD against CLF is yet unclear. Thus, CLF rats transplanted into rat bone marrow-derived mesenchymal stem cells with knocked down Numb gene (BMSCNumb-KD) were treated with HQD. Simultaneously, Numb gene knockdown was also performed in WB-F344 cell line and then treated with refined HQD in vitro. In vivo study revealed that liver fibrosis was inhibited by HQD plus BMSCNumb-KD treatment, while Hyp content in liver tissue, the gene and protein expression of α-SMA, gene expression of Col I, TNF-α, and TGF-β1 were increased compared to that in HQD group. Furthermore, Notch signal pathway was inhibited by HQD plus BMSCNumb-KD, while the protein expression of Numb was decreased and RBP-Jκ and Hes1 was increased compared to that in HQD group. In vitro, HQD reduced the differentiation of WB-F344 cells into cholangiocyte phenotype, while this effect was attenuated after Numb-knockdown. This study highlights that the absence of hepatic stem cell Numb gene decreases effect of HQD against CLF, which give rise the conclusion that Numb might be a potential target for HQD against CLF.
Collapse
Affiliation(s)
- Wen Xu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China
- Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Yan-Nan Xu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China
- Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Xu Zhang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China
- Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Ying Xu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China
- Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Xun Jian
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China
- Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Jia-Mei Chen
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China
- Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Gao-Feng Chen
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China
- Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Hua Zhang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China
- Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Ping Liu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China.
- Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China.
- E-Institute of Shanghai Municipal Education Commission, Shanghai University of TCM, Shanghai, People's Republic of China.
| | - Yong-Ping Mu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China.
- Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China.
| |
Collapse
|
24
|
Efficacy of dietary odd-chain saturated fatty acid pentadecanoic acid parallels broad associated health benefits in humans: could it be essential? Sci Rep 2020; 10:8161. [PMID: 32424181 PMCID: PMC7235264 DOI: 10.1038/s41598-020-64960-y] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
Dietary odd-chain saturated fatty acids (OCFAs) are present in trace levels in dairy fat and some fish and plants. Higher circulating concentrations of OCFAs, pentadecanoic acid (C15:0) and heptadecanoic acid (C17:0), are associated with lower risks of cardiometabolic diseases, and higher dietary intake of OCFAs is associated with lower mortality. Population-wide circulating OCFA levels, however, have been declining over recent years. Here, we show C15:0 as an active dietary fatty acid that attenuates inflammation, anemia, dyslipidemia, and fibrosis in vivo, potentially by binding to key metabolic regulators and repairing mitochondrial function. This is the first demonstration of C15:0's direct role in attenuating multiple comorbidities using relevant physiological mechanisms at established circulating concentrations. Pairing our findings with evidence that (1) C15:0 is not readily made endogenously, (2) lower C15:0 dietary intake and blood concentrations are associated with higher mortality and a poorer physiological state, and (3) C15:0 has demonstrated activities and efficacy that parallel associated health benefits in humans, we propose C15:0 as a potential essential fatty acid. Further studies are needed to evaluate the potential impact of decades of reduced intake of OCFA-containing foods as contributors to C15:0 deficiencies and susceptibilities to chronic disease.
Collapse
|
25
|
Matsuoka Y, Izumi Y, Takahashi M, Bamba T, Yamada KI. Method for Structural Determination of Lipid-Derived Radicals. Anal Chem 2020; 92:6993-7002. [DOI: 10.1021/acs.analchem.0c00053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Yuta Matsuoka
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1 Ohtemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Yoshihiro Izumi
- AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1 Ohtemachi, Chiyoda-ku, Tokyo 100-0004, Japan
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takeshi Bamba
- AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1 Ohtemachi, Chiyoda-ku, Tokyo 100-0004, Japan
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ken-ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1 Ohtemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
26
|
Asokan SM, Hung TH, Li ZY, Chiang WD, Lin WT. Protein hydrolysate from potato confers hepatic-protection in hamsters against high fat diet induced apoptosis and fibrosis by suppressing Caspase-3 and MMP2/9 and by enhancing Akt-survival pathway. Altern Ther Health Med 2019; 19:283. [PMID: 31653214 PMCID: PMC6814989 DOI: 10.1186/s12906-019-2700-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 09/30/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND A potato protein hydrolysate, APPH is a potential anti-obesity diet ingredient. Since, obesity leads to deterioration of liver function and associated liver diseases, in this study the effect of APPH on high fat diet (HFD) associated liver damages was investigated. METHODS Six week old male hamsters were randomly separated to six groups (n = 8) as control, HFD (HFD fed obese), L-APPH (HFD + 15 mg/kg/day of APPH), M-APPH (HFD + 30 mg/kg/day), H-APPH (HFD + 75 mg/kg/day of APPH) and PB (HFD + 500 mg/kg/day of probucol). HFD fed hamsters were administered with APPH 50 days through oral gavage. The animals were euthanized and the number of apoptotic nuclei in liver tissue was determined by TUNEL staining and the extent of interstitial fibrosis was determined by Masson's trichrome staining. Modulation in the molecular events associated with apoptosis and fibrosis were elucidated from the western blotting analysis of the total protein extracts. RESULTS Hamsters fed with high fat diet showed symptoms of liver damage as measured from serum markers like alanine aminotransferase and aspartate aminotransferase levels. However a 50 day long supplementation of APPH effectively ameliorated the effects of HFD. HFD also modulated the expression of survival and apoptosis proteins in the hamster liver. Further the HFD groups showed elevated levels of fibrosis markers in liver. The increase in fibrosis and apoptosis was correlated with the increase in the levels of phosphorylated extracellular signal-regulated kinases (pERK1/2) revealing a potential role of ERK in the HFD mediated liver damage. However APPH treatment reduced the effect of HFD on the apoptosis and fibrosis markers considerably and provided hepato-protection. CONCLUSION APPH can therefore be considered as an efficient therapeutic agent to ameliorate high fat diet related liver damages.
Collapse
|
27
|
Increased adiposity by feeding growing rats a high-fat diet results in iron decompartmentalisation. Br J Nutr 2019; 123:1094-1108. [PMID: 32172712 DOI: 10.1017/s0007114519002320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The present study reports the effects of a high-fat (HF) diet of over 8 weeks on the Fe status of growing rats. Tissue Fe levels were analysed by atomic absorption spectrophotometry, and whole-body adiposity was measured by dual-energy X-ray absorptiometry. Histopathology and morphometry of adipose tissue were performed. Liver homogenates were used for measuring ferroportin-1 protein levels by immunoblotting, and transcript levels were used for Fe genes measured by real-time PCR. Tissue Fe pools were fit to a compartmental biokinetic model in which Fe was assessed using fourteen compartments and twenty-seven transfer constants (kj,i from tissue 'i' to tissue 'j') adapted from the International Commission on Radiological Protection (ICRP) 69. Ten kj,i were calculated from the experimental data using non-linear regression, and seventeen were estimated by allometry according to the formula ${k_{i,j}} = a \times {M^b}$. Validation of the model was carried out by comparing predicted and analysed Fe pool sizes in erythrocytes, the liver and the spleen. Body adiposity was negatively associated with serum Fe levels and positively associated with liver Fe stores. An inferred increase in Fe transfer from bone marrow to the liver paralleled higher hepatic Fe concentrations and ferritin heavy-chain mRNA levels in the HF diet-fed animals, suggesting that liver Fe accumulation occurred at least in part due to a favoured liver erythrocyte uptake. If this feeding condition was to be prolonged, impaired Fe decompartmentalisation may occur, ultimately resulting in dysmetabolic Fe overload.
Collapse
|
28
|
Di Mauro S, Scamporrino A, Petta S, Urbano F, Filippello A, Ragusa M, Di Martino MT, Scionti F, Grimaudo S, Pipitone RM, Privitera G, Di Pino A, Scicali R, Valenti L, Dongiovanni P, Fracanzani A, Rabuazzo AM, Craxì A, Purrello M, Purrello F, Piro S. Serum coding and non-coding RNAs as biomarkers of NAFLD and fibrosis severity. Liver Int 2019; 39:1742-1754. [PMID: 31169972 PMCID: PMC6771597 DOI: 10.1111/liv.14167] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 05/28/2019] [Accepted: 05/31/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS In patients with non-alcoholic fatty liver disease (NAFLD), liver biopsy is the gold standard to detect non-alcoholic steatohepatitis (NASH) and stage liver fibrosis. We aimed to identify differentially expressed mRNAs and non-coding RNAs in serum samples of biopsy-diagnosed mild and severe NAFLD patients with respect to controls and to each other. METHODS We first performed a whole transcriptome analysis through microarray (n = 12: four Control: CTRL; four mild NAFLD: NAS ≤ 4 F0; four severe NAFLD NAS ≥ 5 F3), followed by validation of selected transcripts through real-time PCRs in an independent internal cohort of 88 subjects (63 NAFLD, 25 CTRL) and in an external cohort of 50 NAFLD patients. A similar analysis was also performed on liver biopsies and HepG2 cells exposed to oleate:palmitate or only palmitate (cellular model of NAFL/NASH) at intracellular/extracellular levels. Transcript correlation with histological/clinical data was also analysed. RESULTS We identified several differentially expressed coding/non-coding RNAs in each group of the study cohort. We validated the up-regulation of UBE2V1, BNIP3L mRNAs, RP11-128N14.5 lncRNA, TGFB2/TGFB2-OT1 coding/lncRNA in patients with NAS ≥ 5 (vs NAS ≤ 4) and the up-regulation of HBA2 mRNA, TGFB2/TGFB2-OT1 coding/lncRNA in patients with Fibrosis stages = 3-4 (vs F = 0-2). In in vitro models: UBE2V1, RP11-128N14.5 and TGFB2/TGFB2-OT1 had an increasing expression trend ranging from CTRL to oleate:palmitate or only palmitate-treated cells both at intracellular and extracellular level, while BNIP3L was up-regulated only at extracellular level. UBE2V1, RP11-128N14.5, TGFB2/TGFB2-OT1 and HBA2 up-regulation was also observed at histological level. UBE2V1, RP11-128N14.5, BNIP3L and TGFB2/TGFB2-OT1 correlated with histological/biochemical data. Combinations of TGFB2/TGFB2-OT1 + Fibrosis Index based on the four factors (FIB-4) showed an Area Under the Curve (AUC) of 0.891 (P = 3.00E-06) or TGFB2/TGFB2-OT1 + Fibroscan (AUC = 0.892, P = 2.00E-06) improved the detection of F = 3-4 with respect to F = 0-2 fibrosis stages. CONCLUSIONS We identified specific serum coding/non-coding RNA profiles in severe and mild NAFLD patients that possibly mirror the molecular mechanisms underlying NAFLD progression towards NASH/fibrosis. TGFB2/TGFB2-OT1 detection improves FIB-4/Fibroscan diagnostic performance for advanced fibrosis discrimination.
Collapse
Affiliation(s)
- Stefania Di Mauro
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi‐Nesima HospitalUniversity of CataniaCataniaItaly
| | - Alessandra Scamporrino
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi‐Nesima HospitalUniversity of CataniaCataniaItaly
| | - Salvatore Petta
- Section of Gastroenterology, Di.Bi.M.I.SUniversity of PalermoPalermoItaly
| | - Francesca Urbano
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi‐Nesima HospitalUniversity of CataniaCataniaItaly
| | - Agnese Filippello
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi‐Nesima HospitalUniversity of CataniaCataniaItaly
| | - Marco Ragusa
- Department of BioMedical Sciences and BioTechnologySection of Biology and Genetics Giovanni Sichel, Unit of Molecular, Genome and Complex Systems BioMedicineCataniaItaly,Oasi Research Institute - IRCCSTroina94018Italy
| | - Maria T. Di Martino
- Department of Experimental and Clinical MedicineMagna Graecia UniversityCatanzaroItaly
| | - Francesca Scionti
- Department of Experimental and Clinical MedicineMagna Graecia UniversityCatanzaroItaly
| | - Stefania Grimaudo
- Section of Gastroenterology, Di.Bi.M.I.SUniversity of PalermoPalermoItaly
| | | | - Graziella Privitera
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi‐Nesima HospitalUniversity of CataniaCataniaItaly
| | - Antonino Di Pino
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi‐Nesima HospitalUniversity of CataniaCataniaItaly
| | - Roberto Scicali
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi‐Nesima HospitalUniversity of CataniaCataniaItaly
| | - Luca Valenti
- Translational MedicineUniversity of Milan, Fondazione IRCCS Ca' Granda Pad MarangoniMilanItaly
| | - Paola Dongiovanni
- Department of Pathophysiology and Transplantation, Section of Internal MedicineUniversity of Milan, Fondazione Ca' Granda IRCCS Ospedale Maggiore PoliclinicoMilanItaly
| | - Anna Fracanzani
- Department of Pathophysiology and Transplantation, Section of Internal MedicineUniversity of Milan, Fondazione Ca' Granda IRCCS Ospedale Maggiore PoliclinicoMilanItaly
| | - Agata M. Rabuazzo
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi‐Nesima HospitalUniversity of CataniaCataniaItaly
| | - Antonio Craxì
- Section of Gastroenterology, Di.Bi.M.I.SUniversity of PalermoPalermoItaly
| | - Michele Purrello
- Department of BioMedical Sciences and BioTechnologySection of Biology and Genetics Giovanni Sichel, Unit of Molecular, Genome and Complex Systems BioMedicineCataniaItaly
| | - Francesco Purrello
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi‐Nesima HospitalUniversity of CataniaCataniaItaly
| | - Salvatore Piro
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi‐Nesima HospitalUniversity of CataniaCataniaItaly
| |
Collapse
|
29
|
Yamauchi A, Kamiyoshi A, Sakurai T, Miyazaki H, Hirano E, Lim HS, Kaku T, Shindo T. Development of a mouse iron overload-induced liver injury model and evaluation of the beneficial effects of placenta extract on iron metabolism. Heliyon 2019; 5:e01637. [PMID: 31193082 PMCID: PMC6515419 DOI: 10.1016/j.heliyon.2019.e01637] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/16/2019] [Accepted: 04/30/2019] [Indexed: 12/11/2022] Open
Abstract
Hepatic iron deposition is seen in cases of chronic hepatitis and cirrhosis, and is a hallmark of a poorer prognosis. Iron deposition is also found in non-alcoholic steatohepatitis (NASH) patients. We have now developed a mouse model of NASH with hepatic iron deposition by combining a methione- and choline-deficient (MCD) diet with an iron-overload diet. Using this model, we evaluated the effects of human placenta extract (HPE), which has been shown to ameliorate the pathology of NASH. Four-week-old male C57BL/6 mice were fed the MCD diet with 2% iron for 12 weeks. In liver sections, iron deposition was first detected around the portal vein after 1 week. From there it spread throughout the parenchyma. Biliary iron concentrations were continuously elevated throughout the entire 12-week diet. As a compensatory response, the diet caused elevation of serum hepcidin, which accelerates excretion of iron from the body. Accumulation of F4/80-positive macrophages was detected within the sinusoids from the first week onward, and real-time PCR analysis revealed elevated hepatic expression of genes related inflammation and oxidative stress. In the model mice, HPE treatment led to a marked reduction of hepatic iron deposition with a corresponding increase in biliary iron excretion. Macrophage accumulation was much reduced by HPE treatment, as was the serum oxidation-reduction potential, an index of oxidative stress. These data indicate that by suppressing inflammation, oxidative stress and iron deposition, and enhancing iron excretion, HPE effectively ameliorates iron overload-induced liver injury. HPE administration may thus be an effective strategy for treating NASH.
Collapse
Affiliation(s)
- Akihiro Yamauchi
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Matsumoto, Japan.,Japan Bio Products Co., Ltd., Tokyo, Japan
| | - Akiko Kamiyoshi
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Takayuki Sakurai
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | | | | | | | | | - Takayuki Shindo
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| |
Collapse
|
30
|
Xu W, Peng F, Deng Y, Fan X, Li N. The emerging roles of eryptosis in liver diseases. Transfus Clin Biol 2019; 26:336-340. [PMID: 31201023 DOI: 10.1016/j.tracli.2019.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/23/2019] [Indexed: 12/16/2022]
Abstract
Erythrocytes undergo programmed cell death, similar to apoptosis, known as eryptosis. This process is a result of several factors including hyperosmolarity, oxidative stress, and exposure to xenobiotics, and is characterized by the breakdown of membrane phospholipid asymmetry, the clustering of band 3, and the generation of red blood cell-derived microparticles. Under pathological conditions, the liver is the primary site of erythrocyte clearance and plays an important role in iron recycling. Phosphatidylserine exposure and band-3 clustering on eryptotic erythrocytes represent mainly pro-phagocytic signals. Further, the percentage of eryptotic erythrocytes is enhanced in the circulating blood of patients with hepatic failure, hyperbilirubinemia, and nonalcoholic steatohepatitis. In this review, we concentrate on recent progress regarding the pathophysiological roles of eryptosis in liver diseases.
Collapse
Affiliation(s)
- Wei Xu
- Department of Blood Transfusion, Central South University, Xiangya Hospital, 410008 Changsha, China
| | - Fang Peng
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Central South University, Xiangya Hospital, 410008 Changsha, China
| | - Ying Deng
- The Hospital of Ningxiang County People, 410600 Changsha, China
| | - Xuegong Fan
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan Province, Central South University, Xiangya Hospital, 410008 Changsha, China
| | - Ning Li
- Department of Blood Transfusion, Central South University, Xiangya Hospital, 410008 Changsha, China.
| |
Collapse
|
31
|
Iron-Induced Liver Injury: A Critical Reappraisal. Int J Mol Sci 2019; 20:ijms20092132. [PMID: 31052166 PMCID: PMC6539962 DOI: 10.3390/ijms20092132] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/25/2019] [Accepted: 04/27/2019] [Indexed: 12/12/2022] Open
Abstract
Iron is implicated in the pathogenesis of a number of human liver diseases. Hereditary hemochromatosis is the classical example of a liver disease caused by iron, but iron is commonly believed to contribute to the progression of other forms of chronic liver disease such as hepatitis C infection and nonalcoholic fatty liver disease. In this review, we present data from cell culture experiments, animal models, and clinical studies that address the hepatotoxicity of iron. These data demonstrate that iron overload is only weakly fibrogenic in animal models and rarely causes serious liver damage in humans, calling into question the concept that iron overload is an important cause of hepatotoxicity. In situations where iron is pathogenic, iron-induced liver damage may be potentiated by coexisting inflammation, with the resulting hepatocyte necrosis an important factor driving the fibrogenic response. Based on the foregoing evidence that iron is less hepatotoxic than is generally assumed, claims that assign a causal role to iron in liver injury in either animal models or human liver disease should be carefully evaluated.
Collapse
|
32
|
Dybas J, Grosicki M, Baranska M, Marzec KM. Raman imaging of heme metabolism in situ in macrophages and Kupffer cells. Analyst 2019; 143:3489-3498. [PMID: 29951676 DOI: 10.1039/c8an00282g] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Herein, we provide the Raman imaging results for different stages of erythrophagocytosis of senescent red blood cells executed by isolated murine primary Kupffer cells and a murine macrophage cell line (RAW 264.7). Images were recorded with the use of 488 and 532 nm excitation lines. The use of Resonance Raman spectroscopy allowed the visualization of the heme metabolism and tracking of the systemic iron recycling process inside isolated murine Kupffer cells and RAW.264 cells. Because of the application of the different experimental assays, the erythrophagocytosis in two types of cells was presented and associated with the presence of different forms of oxidized and degradative derivatives of hemoglobin species. Moreover, we observed an increase of lipid level and later formation of lipid droplets during the erythrophagocytosis process inside RAW 264.7 macrophages and murine Kupffer cells.
Collapse
Affiliation(s)
- J Dybas
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), 14 Bobrzyńskiego Str., 30-348 Krakow, Poland.
| | | | | | | |
Collapse
|
33
|
The role of macrophages in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Nat Rev Gastroenterol Hepatol 2019; 16:145-159. [PMID: 30482910 DOI: 10.1038/s41575-018-0082-x] [Citation(s) in RCA: 647] [Impact Index Per Article: 107.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and its inflammatory and often progressive subtype nonalcoholic steatohepatitis (NASH) are becoming the leading cause of liver-related morbidity and mortality worldwide, and a primary indication for liver transplantation. The pathophysiology of NASH is multifactorial and not yet completely understood; however, innate immunity is a major contributing factor in which liver-resident macrophages (Kupffer cells) and recruited macrophages play a central part in disease progression. In this Review, we assess the evidence for macrophage involvement in the development of steatosis, inflammation and fibrosis in NASH. In this process, not only the polarization of liver macrophages towards a pro-inflammatory phenotype is important, but adipose tissue macrophages, especially in the visceral compartment, also contribute to disease severity and insulin resistance. Macrophage activation is mediated by factors such as endotoxins and translocated bacteria owing to increased intestinal permeability, factors released from damaged or lipoapoptotic hepatocytes, as well as alterations in gut microbiota and defined nutritional components, including certain free fatty acids, cholesterol and their metabolites. Reflecting the important role of macrophages in NASH, we also review studies investigating drugs that target macrophage recruitment to the liver, macrophage polarization and their inflammatory effects as potential treatment options for patients with NASH.
Collapse
|
34
|
Zhao QH, Zhang XS, Wu K, Zhang J, Xia TF, Chen J, Qin ZS, Pang LQ. Preparation of Zoledronate liposome and its impact on apoptosis of Kupffer cells in rat liver. Acta Cir Bras 2019; 33:1052-1060. [PMID: 30624510 DOI: 10.1590/s0102-865020180120000002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/16/2018] [Indexed: 01/07/2023] Open
Abstract
PURPOSE To establish a method for the preparation of zoledronate liposome and to observe its effect on inducing the apoptosis of rat liver Kupffer cells. METHODS Zoledronate was encapsulated in liposomes, and then the entrapment rate was detected on a spectrophotometer. The prepared Zoledronate liposome (0.01 mg/mL) was injected into the tail vein of SD rats. Three days later, the number of Kupffer cells (CD68 positive) in rat liver tissue was detected by immunohistochemistry. Flow cytometry was used to detect the apoptosis rate of the isolated liver Kupffer cell cultured in vitro. RESULTS The entrapment rate of Zoledronate was 43.4±7.8%. Immunohistochemistry revealed that the number of Kupffer cells was 19.3±2.1 in PBS group and 5.5±1.7 in Zoledronate liposome group, with a significant difference (P<0.05). The apoptosis rate of Kupffer cells was 4.1±0.8% in PBS group, while it was 9±2.2% and 23.3±5.9% in Zoledronate liposomes groups with different concentrations of Zoledronate liposome (P<0.05). CONCLUSIONS Zoledronate liposomes can effectively induce the apoptosis of Kupffer cells in vivo and in vitro, and the apoptosis rate is related to the concentration of Zoledronate liposome. To establish a rat liver Kupffer cell apoptosis model can provide a new means for further study on Kupffer cell function.
Collapse
Affiliation(s)
- Qiao-Hong Zhao
- Bachelor of Medical Science, Department of Nursing, Jiangsu College of Nursing, Huai'an, China. Conception of the study, acquisition and interpretation of data
| | - Xi-Shan Zhang
- Bachelor of Medical Science, Department of General Surgery, Lian'shui County People's Hospital, Lian'shui, China. Analysis and interpretation of data
| | - Kun Wu
- Fellow Master degree, Postgraduate Program in Surgical Science, Department of General Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China. Immunohistochemical and flow cytometry analysis
| | - Jie Zhang
- Fellow Master degree, Postgraduate Program in Surgical Science, Department of General Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China. Immunohistochemical and flow cytometry analysis
| | - Tian-Fang Xia
- Fellow Master degree, Postgraduate Program in Surgical Science, Department of General Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China. Immunohistochemical and flow cytometry analysis
| | - Jian Chen
- Fellow Master degree, Postgraduate Program in Surgical Science, Department of General Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China. Immunohistochemical and flow cytometry analysis
| | - Zhen-Shen Qin
- PhD, Associate Professor, Department of General Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China. Conception and design of the study, critical revision
| | - Li-Qun Pang
- PhD, Associate Professor, Department of General Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China. Conception and design of the study, critical revision
| |
Collapse
|
35
|
Thuy LTT, Hai H, Hieu VN, Dat NQ, Hoang DV, Kawada N. Antifibrotic Therapy for Liver Cirrhosis. THE EVOLVING LANDSCAPE OF LIVER CIRRHOSIS MANAGEMENT 2019:167-189. [DOI: 10.1007/978-981-13-7979-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
36
|
Neave MJ, Hall RN, Huang N, McColl KA, Kerr P, Hoehn M, Taylor J, Strive T. Robust Innate Immunity of Young Rabbits Mediates Resistance to Rabbit Hemorrhagic Disease Caused by Lagovirus Europaeus GI.1 But Not GI.2. Viruses 2018; 10:E512. [PMID: 30235853 PMCID: PMC6163550 DOI: 10.3390/v10090512] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/12/2018] [Accepted: 09/15/2018] [Indexed: 01/07/2023] Open
Abstract
The rabbit caliciviruses Lagovirus europaeus GI.1 and GI.2 both cause acute necrotizing hepatitis in European rabbits (Oryctolagus cuniculus). Whilst GI.2 is highly virulent in both young and adult rabbits, rabbits younger than eight weeks of age are highly resistant to disease caused by GI.1, although they are still permissive to infection and viral replication. To investigate the underlying mechanism(s) of this age related resistance to GI.1, we compared liver transcriptomes of young rabbits infected with GI.1 to those of adult rabbits infected with GI.1 and young rabbits infected with GI.2. Our data suggest that kittens have constitutively heightened innate immune responses compared to adult rabbits, particularly associated with increased expression of major histocompatibility class II molecules and activity of natural killer cells, macrophages, and cholangiocytes. This enables them to respond more rapidly to GI.1 infection than adult rabbits and thus limit virus-induced pathology. In contrast, these responses were not fully developed during GI.2 infection. We speculate that the observed downregulation of multiple genes associated with innate immunity in kittens during GI.2 infection may be due to virally-mediated immunomodulation, permitting fatal disease to develop. Our study provides insight into the fundamental host⁻pathogen interactions responsible for the differences in age-related susceptibility, which likely plays a critical role in defining the success of GI.2 in outcompeting GI.1 in the field.
Collapse
Affiliation(s)
- Matthew J Neave
- CSIRO Australian Animal Health Laboratory, Geelong, VIC 3220, Australia.
| | - Robyn N Hall
- CSIRO Health and Biosecurity, Canberra, ACT 2601, Australia.
| | - Nina Huang
- CSIRO Health and Biosecurity, Canberra, ACT 2601, Australia.
| | - Kenneth A McColl
- CSIRO Australian Animal Health Laboratory, Geelong, VIC 3220, Australia.
| | - Peter Kerr
- CSIRO Health and Biosecurity, Canberra, ACT 2601, Australia.
| | - Marion Hoehn
- CSIRO Health and Biosecurity, Canberra, ACT 2601, Australia.
| | | | - Tanja Strive
- CSIRO Health and Biosecurity, Canberra, ACT 2601, Australia.
| |
Collapse
|
37
|
Masarone M, Rosato V, Dallio M, Gravina AG, Aglitti A, Loguercio C, Federico A, Persico M. Role of Oxidative Stress in Pathophysiology of Nonalcoholic Fatty Liver Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9547613. [PMID: 29991976 PMCID: PMC6016172 DOI: 10.1155/2018/9547613] [Citation(s) in RCA: 468] [Impact Index Per Article: 66.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/23/2018] [Indexed: 02/06/2023]
Abstract
Liver steatosis without alcohol consumption, namely, nonalcoholic fatty liver disease (NAFLD), is a common hepatic condition that encompasses a wide spectrum of presentations, ranging from simple accumulation of triglycerides in the hepatocytes without any liver damage to inflammation, necrosis, ballooning, and fibrosis (namely, nonalcoholic steatohepatitis) up to severe liver disease and eventually cirrhosis and/or hepatocellular carcinoma. The pathophysiology of fatty liver and its progression is influenced by multiple factors (environmental and genetics), in a "multiple parallel-hit model," in which oxidative stress plays a very likely primary role as the starting point of the hepatic and extrahepatic damage. The aim of this review is to give a comprehensive insight on the present researches and findings on the role of oxidative stress mechanisms in the pathogenesis and pathophysiology of NAFLD. With this aim, we evaluated the available data in basic science and clinical studies in this field, reviewing the most recent works published on this topic.
Collapse
Affiliation(s)
- Mario Masarone
- Internal Medicine and Hepatology Division, Department of Medicine, University of Medicine of Salerno, Salerno, Italy
| | - Valerio Rosato
- Internal Medicine and Hepatology Division, Department of Medicine, University of Medicine of Salerno, Salerno, Italy
| | - Marcello Dallio
- Hepatogastroenterology Division, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Antonietta Gerarda Gravina
- Internal Medicine and Hepatology Division, Department of Medicine, University of Medicine of Salerno, Salerno, Italy
| | - Andrea Aglitti
- Internal Medicine and Hepatology Division, Department of Medicine, University of Medicine of Salerno, Salerno, Italy
| | - Carmelina Loguercio
- Hepatogastroenterology Division, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Alessandro Federico
- Hepatogastroenterology Division, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Marcello Persico
- Internal Medicine and Hepatology Division, Department of Medicine, University of Medicine of Salerno, Salerno, Italy
| |
Collapse
|
38
|
Al-Quraishy S, Dkhil MA, Al-Shaebi EM, Abdel-Baki AAS, Araúzo-Bravo MJ, Delic D, Wunderlich F. Gene expression of the liver of vaccination-protected mice in response to early patent infections of Plasmodium chabaudi blood-stage malaria. Malar J 2018; 17:215. [PMID: 29843710 PMCID: PMC5975554 DOI: 10.1186/s12936-018-2366-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/23/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The role of the liver for survival of blood-stage malaria is only poorly understood. In experimental blood-stage malaria with Plasmodium chabaudi, protective vaccination induces healing and, thus, survival of otherwise lethal infections. This model is appropriate to study the role of the liver in vaccination-induced survival of blood-stage malaria. METHODS Female Balb/c mice were vaccinated with a non-infectious vaccine consisting of plasma membranes isolated in the form of erythrocyte ghosts from P. chabaudi-infected erythrocytes at week 3 and week 1 before infection with P. chabaudi blood-stage malaria. Gene expression microarrays and quantitative real-time PCR were used to investigate the response of the liver, in terms of expression of mRNA and long intergenic non-coding (linc)RNA, to vaccination-induced healing infections and lethal P. chabaudi malaria at early patency on day 4 post infection, when parasitized erythrocytes begin to appear in peripheral blood. RESULTS In vaccination-induced healing infections, 23 genes were identified to be induced in the liver by > tenfold at p < 0.01. More than one-third were genes known to be involved in erythropoiesis, such as Kel, Rhag, Ahsp, Ermap, Slc4a1, Cldn13 Gata1, and Gfi1b. Another group of > tenfold expressed genes include genes involved in natural cytotoxicity, such as those encoding killer cell lectin-like receptors Klrb1a, Klrc3, Klrd1, the natural cytotoxicity-triggering receptor 1 Ncr1, as well as the granzyme B encoding Gzmb. Additionally, a series of genes involved in the control of cell cycle and mitosis were identified: Ccnb1, Cdc25c, Ckap2l were expressed > tenfold only in vaccination-protected mice, and the expression of 22 genes was at least 100% higher in vaccination-protected mice than in non-vaccinated mice. Furthermore, distinct lincRNA species were changed by > threefold in livers of vaccination-protected mice, whereas lethal malaria induced different lincRNAs. CONCLUSION The present data suggest that protective vaccination accelerates the malaria-induced occurrence of extramedullary erythropoiesis, generation of liver-resident cytotoxic cells, and regeneration from malaria-induced injury in the liver at early patency, which may be critical for final survival of otherwise lethal blood-stage malaria of P. chabaudi.
Collapse
Affiliation(s)
- Saleh Al-Quraishy
- Department of Zoology, College of Science, King Saud University, P.O. Box: 2455, Riyadh, 11451, Saudi Arabia
| | - Mohamed A Dkhil
- Department of Zoology, College of Science, King Saud University, P.O. Box: 2455, Riyadh, 11451, Saudi Arabia.
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt.
| | - E M Al-Shaebi
- Department of Zoology, College of Science, King Saud University, P.O. Box: 2455, Riyadh, 11451, Saudi Arabia
| | - Abdel-Azeem S Abdel-Baki
- Department of Zoology, College of Science, King Saud University, P.O. Box: 2455, Riyadh, 11451, Saudi Arabia
- Department of Zoology, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Marcos J Araúzo-Bravo
- Group of Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, San Sebastián, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Denis Delic
- Boehringer-Ingelheim Pharma, Biberach, Germany
| | - Frank Wunderlich
- Department of Biology, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
39
|
Al-Quraishy S, Dkhil MA, Abdel-Baki AAS, Delic D, Wunderlich F. Protective vaccination alters gene expression of the liver of Balb/c mice in response to early prepatent blood-stage malaria of Plasmodium chabaudi. Parasitol Res 2018; 117:1115-1129. [DOI: 10.1007/s00436-018-5789-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/25/2018] [Indexed: 12/19/2022]
|
40
|
Machado MV, Diehl AM. Pathogenesis of Nonalcoholic Fatty Liver Disease. ZAKIM AND BOYER'S HEPATOLOGY 2018:369-390.e14. [DOI: 10.1016/b978-0-323-37591-7.00025-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
41
|
Matsushita N, Hassanein MT, Martinez-Clemente M, Lazaro R, French SW, Xie W, Lai K, Karin M, Tsukamoto H. Gender difference in NASH susceptibility: Roles of hepatocyte Ikkβ and Sult1e1. PLoS One 2017; 12:e0181052. [PMID: 28797077 PMCID: PMC5552280 DOI: 10.1371/journal.pone.0181052] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 06/26/2017] [Indexed: 12/22/2022] Open
Abstract
Myeloid cell and hepatocyte IKKβ may mediate the genesis of obesity and insulin resistance in mice fed high fat diet. However, their gender-specific roles in the pathogenesis of non-alcoholic steatohepatitis (NASH) are not known. Here we demonstrate myeloid IKKβ deficiency prevents Western diet-induced obesity and visceral adiposity in females but not in males, and attenuates hyperglycemia, global IR, and NASH in both genders. In contrast, all metabolic sequela including NASH are aggravated by hepatocyte IKKβ deficiency (IkbkbΔhep) in male but not female mice. Gene profiling identifies sulfotransferase family 1E (Sult1e1), which encodes a sulfotransferase E1 responsible for inactivation of estrogen, as a gene upregulated in NASH in both genders and most conspicuously in male IkbkbΔhep mice having worst NASH and lowest plasma estradiol levels. LXRα is enriched to LXRE on Sult1e1 promoter in male WT and IkbkbΔhep mice with NASH, and a Sult1e1 promoter activity is increased by LXRα and its ligand and augmented by expression of a S32A mutant of IκBα. These results demonstrate striking gender differences in regulation by IKKβ of high cholesterol saturated fat diet-induced metabolic changes including NASH and suggest hepatocyte IKKβ is protective in male due at least in part to its ability to repress LXR-induced Sult1e1. Our findings also raise a caution for systemic IKK inhibition for the treatment of NASH as it may exacerbate the disease in male patients.
Collapse
Affiliation(s)
- Noriko Matsushita
- Southern California Research Center for ALPD and Cirrhosis of the University of Southern California, Los Angeles, California, United States of America
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Mohamed T. Hassanein
- Southern California Research Center for ALPD and Cirrhosis of the University of Southern California, Los Angeles, California, United States of America
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Marcos Martinez-Clemente
- Southern California Research Center for ALPD and Cirrhosis of the University of Southern California, Los Angeles, California, United States of America
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Raul Lazaro
- Southern California Research Center for ALPD and Cirrhosis of the University of Southern California, Los Angeles, California, United States of America
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Samuel W. French
- Southern California Research Center for ALPD and Cirrhosis of the University of Southern California, Los Angeles, California, United States of America
- Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Wen Xie
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, United States of America
| | - Keane Lai
- Southern California Research Center for ALPD and Cirrhosis of the University of Southern California, Los Angeles, California, United States of America
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Michael Karin
- Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, California, United States of America
| | - Hidekazu Tsukamoto
- Southern California Research Center for ALPD and Cirrhosis of the University of Southern California, Los Angeles, California, United States of America
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
- Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
42
|
Chen J, Li X, Hu Y, Liu W, Zhou Q, Zhang H, Mu Y, Liu P. Gypenosides Ameliorate Carbon Tetrachloride-Induced Liver Fibrosis by Inhibiting the Differentiation of Hepatic Progenitor Cells into Myofibroblasts. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:1061-1074. [PMID: 28659031 DOI: 10.1142/s0192415x17500574] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Gypenosides (GPs), the predominant components of Gynostemma pentaphyllum, exert antifibrotic effects; however, the mechanisms underlying their ability to ameliorate liver fibrosis are unclear. Liver fibrosis was induced in C57BL/6 mice via subcutaneous injection of 10% carbon tetrachloride (CCl[Formula: see text] three times a week for two weeks. Then, CCl4 was administered in conjunction with intragastric GPs for another three weeks. For in vitro analyses, WB-F344, hepatatic progenitor cells (HPCs) were treated with transforming growth factor beta 1 (TGF-[Formula: see text]1) with or without GPs for 48[Formula: see text]h. The results showed that alanine aminotransferase (ALT) and aspartate transaminase (AST) activity, deposition of collagen, hydroxyproline content, and expression of alpha-smooth muscle actin ([Formula: see text]-SMA) and collagen type I (Col I) were significantly decreased after treatment with GPs ([Formula: see text], [Formula: see text], [Formula: see text], [Formula: see text]). In the 5M CCl4 group, the expression of HPC markers, Sox9 and cytokeratin 19 (CK19), was significantly increased compared with the normal or GPs-treated group ([Formula: see text], [Formula: see text]). Immunostaining showed that the number of Sox9 and [Formula: see text]-SMA double-positive cells was higher in the 5M CCl4 group than in the normal group, but the addition of GPs caused this cell number to decrease. In WB-F344 cells, the expression of [Formula: see text]-SMA and Col I was significantly increased after treatment with TGF-[Formula: see text], whereas in the GPs treatment group, expression was markedly decreased ([Formula: see text]). The levels of TGF-[Formula: see text] and TGF-[Formula: see text]R1 were markedly reduced after GPs treatment both in vivo and in vitro. In conclusion, GPs ameliorated CCl4-induced liver fibrosis via the inhibition of TGF-[Formula: see text] signaling, consequently inhibiting the differentiation of HPCs into myofibroblasts.
Collapse
Affiliation(s)
- Jiamei Chen
- * Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- † Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xuewei Li
- * Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- † Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yonghong Hu
- * Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- † Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wei Liu
- * Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- † Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qun Zhou
- * Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- † Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hua Zhang
- * Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- † Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yongping Mu
- * Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- † Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ping Liu
- * Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- † Shanghai Key Laboratory of Traditional Chinese Medicine, Shanghai 201203, China
- ‡ E-Institute of Shanghai Municipal Education Commission, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
43
|
Cheon SY, Chung KS, Lee KJ, Choi HY, Ham IH, Jung DH, Cha YY, An HJ. HVC1 ameliorates hyperlipidemia and inflammation in LDLR -/- mice. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:222. [PMID: 28427397 PMCID: PMC5397752 DOI: 10.1186/s12906-017-1734-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 04/08/2017] [Indexed: 12/29/2022]
Abstract
Background HVC1 consists of Coptidis Rhizoma (dried rhizome of Coptischinensis), Scutellariae Radix (root of Scutellariabaicalensis), Rhei Rhizoma (rhizome of Rheum officinale), and Pruni Cortex (cortex of Prunusyedoensis Matsum). Although the components are known to be effective in various conditions such as inflammation, hypertension, and hypercholesterolemia, there are no reports of the molecular mechanism of its hypolipidemic effects. Methods We investigated the hypolipidemic effect of HVC1 in low-density lipoprotein receptor-deficient (LDLR−/−) mice fed a high-cholesterol diet for 13 weeks. Mice were randomized in to 6 groups: ND (normal diet) group, HCD (high-cholesterol diet) group, and treatment groups fed HCD and treated with simvastatin (10 mg/kg, p.o.) or HVC1 (10, 50, or 250 mg/kg, p.o.). Results HVC1 regulated the levels of total cholesterol, triglyceride (TG), low-density lipoprotein (LDL) cholesterol, and high-density lipoprotein (HDL) cholesterol in mouse serum. In addition, it regulated the transcription level of the peroxisome proliferator-activated receptors (PPARs), sterol regulatory element-binding proteins (SREBP)-2, 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase, lipoprotein lipase (LPL), apolipoprotein B (apo B), liver X receptor (LXR), and inflammatory cytokines (IL-1β, IL-6, and TNF-α). Furthermore, HVC1 activated 5′ adenosine monophosphate-activated protein kinase (AMPK). Conclusion Our results suggest that HVC1 might be effective in preventing high-cholesterol diet-induced hyperlipidemia by regulating the genes involved in cholesterol and lipid metabolism, and inflammatory responses.
Collapse
|
44
|
Protective vaccination and blood-stage malaria modify DNA methylation of gene promoters in the liver of Balb/c mice. Parasitol Res 2017; 116:1463-1477. [PMID: 28315013 DOI: 10.1007/s00436-017-5423-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/08/2017] [Indexed: 02/07/2023]
Abstract
Epigenetic mechanisms such as DNA methylation are increasingly recognized to be critical for vaccination efficacy and outcome of different infectious diseases, but corresponding information is scarcely available for host defense against malaria. In the experimental blood-stage malaria Plasmodium chabaudi, we investigate the possible effects of a blood-stage vaccine on DNA methylation of gene promoters in the liver, known as effector against blood-stage malaria, using DNA methylation microarrays. Naturally susceptible Balb/c mice acquire, by protective vaccination, the potency to survive P. chabaudi malaria and, concomitantly, modifications of constitutive DNA methylation of promoters of numerous genes in the liver; specifically, promoters of 256 genes are hyper(=up)- and 345 genes are hypo(=down)-methylated (p < 0.05). Protective vaccination also leads to changes in promoter DNA methylation upon challenge with P. chabaudi at peak parasitemia on day 8 post infection (p.i.), when 571 and 1013 gene promoters are up- and down-methylated, respectively, in relation to constitutive DNA methylation (p < 0.05). Gene set enrichment analyses reveal that both vaccination and P. chabaudi infections mainly modify promoters of those genes which are most statistically enriched with functions relating to regulation of transcription. Genes with down-methylated promoters encompass those encoding CX3CL1, GP130, and GATA2, known to be involved in monocyte recruitment, IL-6 trans-signaling, and onset of erythropoiesis, respectively. Our data suggest that vaccination may epigenetically improve parts of several effector functions of the liver against blood-stage malaria, as, e.g., recruitment of monocyte/macrophage to the liver accelerated liver regeneration and extramedullary hepatic erythropoiesis, thus leading to self-healing of otherwise lethal P. chabaudi blood-stage malaria.
Collapse
|
45
|
Zhang X, Xu Y, Chen JM, Liu C, Du GL, Zhang H, Chen GF, Jiang SL, Liu CH, Mu YP, Liu P. Huang Qi Decoction Prevents BDL-Induced Liver Fibrosis Through Inhibition of Notch Signaling Activation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:85-104. [PMID: 28081630 DOI: 10.1142/s0192415x17500070] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Notch signaling has been demonstrated to be involved in ductular reactions and fibrosis. Previous studies have shown that Huang Qi Decoction (HQD) can prevent the progression of cholestatic liver fibrosis (CLF). However, whether HQD affects the Notch signaling pathway is unclear. In this study, CLF was established by common bile duct ligation (BDL) in rats. At the end of the first week, the rats were randomly divided into a model group (i.e., BDL), an HQD group, and a sorafenib positive control group (SORA) and were treated for 3 weeks. Bile duct proliferation and liver fibrosis were determined by tissue staining. Activation of the Notch signaling pathway was evaluated by analyzing expressions of Notch-1, -2, -3, and -4, Jagged (JAG) 1, and Delta like (DLL)-1, -3, and -4. The results showed that HQD significantly reduced the deposition of collagen and the Hyp content of liver tissue and inhibited the activation of HSCs compared with the BDL group. In addition, HQD significantly decreased the protein and mRNA expressions of TGF-[Formula: see text]1 and [Formula: see text]-SMA. In contrast, HQD significantly enhanced expression of the Smad 7 protein. HQD also reduced biliary epithelial cell proliferation, and reduced the mRNA levels of CK7, CK8, CK18, SRY-related high mobility group-box gene (SOX) 9, epithelial cell adhesion molecule (EpCAM) and the positive areas of CK19 and OV6. In addition, the mRNA and protein expressions of Notch-3, -4, JAG1, and DLL-1, -3 were significantly reduced in the HQD compared to the BDL group. These results demonstrated that HQD may prevent biliary liver fibrosis through inhibition of the Notch signaling pathway, and it may be a potential treatment for cholestatic liver disease.
Collapse
Affiliation(s)
- Xiao Zhang
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
- § Longhua Hospital Affiliated to Shanghai University of TCM, Shanghai, P.R. China
| | - Ying Xu
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Jia-Mei Chen
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Cheng Liu
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Guang-Li Du
- † E-institute of Shanghai Municipal Education Commission, Shanghai University of TCM, Shanghai, P.R. China
| | - Hua Zhang
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Gao-Feng Chen
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Shi-Li Jiang
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Cheng-Hai Liu
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Yong-Ping Mu
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Ping Liu
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- † E-institute of Shanghai Municipal Education Commission, Shanghai University of TCM, Shanghai, P.R. China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| |
Collapse
|
46
|
Dkhil MA, Al-Quraishy SA, Abdel-Baki AAS, Delic D, Wunderlich F. Differential miRNA Expression in the Liver of Balb/c Mice Protected by Vaccination during Crisis of Plasmodium chabaudi Blood-Stage Malaria. Front Microbiol 2017; 7:2155. [PMID: 28123381 PMCID: PMC5225092 DOI: 10.3389/fmicb.2016.02155] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/21/2016] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs are increasingly recognized as epigenetic regulators for outcome of diverse infectious diseases and vaccination efficacy, but little information referring to this exists for malaria. This study investigates possible effects of both protective vaccination and P. chabaudi malaria on the miRNome of the liver as an effector against blood-stage malaria using miRNA microarrays and quantitative PCR. Plasmodium chabaudi blood-stage malaria takes a lethal outcome in female Balb/c mice, but a self-healing course after immunization with a non-infectious blood-stage vaccine. The liver robustly expresses 71 miRNA species at varying levels, among which 65 miRNA species respond to malaria evidenced as steadily increasing or decreasing expressions reaching highest or lowest levels toward the end of the crisis phase on day 11 p.i. in lethal malaria. Protective vaccination does not affect constitutive miRNA expression, but leads to significant (p < 0.05) changes in the expression of 41 miRNA species, however evidenced only during crisis. In vaccination-induced self-healing infections, 18 miRNA-species are up- and 14 miRNA-species are down-regulated by more than 50% during crisis in relation to non-vaccinated mice. Vaccination-induced self-healing and survival of otherwise lethal infections of P. chabaudi activate epigenetic miRNA-regulated remodeling processes in the liver manifesting themselves during crisis. Especially, liver regeneration is accelerated as suggested by upregulation of let-7a-5p, let-7b-5p, let-7c-5p, let-7d-5p, let-7f-5p, let-7g-5p, let-7i-5p, miR-26a, miR-122-5p, miR30a, miR27a, and mir-29a, whereas the up-regulated expression of miR-142-3p by more than 100% is compatible with the view of enhanced hepatic erythropoiesis, possibly at expense of megakaryopoiesis, during crisis of P. chabaudi blood-stage malaria.
Collapse
Affiliation(s)
- Mohamed A Dkhil
- Department of Zoology, College of Science, King Saud UniversityRiyadh, Saudi Arabia; Department of Zoology and Entomology, Faculty of Science, Helwan UniversityCairo, Egypt
| | - Saleh A Al-Quraishy
- Department of Zoology, College of Science, King Saud University Riyadh, Saudi Arabia
| | - Abdel-Azeem S Abdel-Baki
- Department of Zoology, College of Science, King Saud UniversityRiyadh, Saudi Arabia; Department of Zoology, Faculty of Science, Beni-Suef UniversityBeni-Suef, Egypt
| | - Denis Delic
- Boehringer-Ingelheim Pharma Biberach, Germany
| | - Frank Wunderlich
- Department of Biology, Heinrich-Heine-University Duesseldorf, Germany
| |
Collapse
|
47
|
Ali RFM, El-Anany AM. Hypolipidemic and Hypocholesterolemic Effect of Roselle ( Hibiscus sabdariffa L.) Seeds Oil in Experimental Male Rats. J Oleo Sci 2017; 66:41-49. [DOI: 10.5650/jos.ess16126] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Rehab F. M. Ali
- Biochemistry Department, Faculty of Agriculture, Cairo University
| | - Ayman M. El-Anany
- Department of Special Food and Nutrition Researches, Food Tech. Res. Institute; Agricultural Research Center
| |
Collapse
|
48
|
Britton LJ, Subramaniam VN, Crawford DHG. Iron and non-alcoholic fatty liver disease. World J Gastroenterol 2016; 22:8112-8122. [PMID: 27688653 PMCID: PMC5037080 DOI: 10.3748/wjg.v22.i36.8112] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/06/2016] [Accepted: 08/05/2016] [Indexed: 02/06/2023] Open
Abstract
The mechanisms that promote liver injury in non-alcoholic fatty liver disease (NAFLD) are yet to be thoroughly elucidated. As such, effective treatment strategies are lacking and novel therapeutic targets are required. Iron has been widely implicated in the pathogenesis of NAFLD and represents a potential target for treatment. Relationships between serum ferritin concentration and NAFLD are noted in a majority of studies, although serum ferritin is an imprecise measure of iron loading. Numerous mechanisms for a pathogenic role of hepatic iron in NAFLD have been demonstrated in animal and cell culture models. However, the human data linking hepatic iron to liver injury in NAFLD is less clear, with seemingly conflicting evidence, supporting either an effect of iron in hepatocytes or within reticulo-endothelial cells. Adipose tissue has emerged as a key site at which iron may have a pathogenic role in NAFLD. Evidence for this comes indirectly from studies that have evaluated the role of adipose tissue iron with respect to insulin resistance. Adding further complexity, multiple strands of evidence support an effect of NAFLD itself on iron metabolism. In this review, we summarise the human and basic science data that has evaluated the role of iron in NAFLD pathogenesis.
Collapse
|
49
|
Chen G, Ni Y, Nagata N, Xu L, Ota T. Micronutrient Antioxidants and Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2016; 17:ijms17091379. [PMID: 27563875 PMCID: PMC5037659 DOI: 10.3390/ijms17091379] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/12/2016] [Accepted: 08/17/2016] [Indexed: 12/14/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most important chronic liver diseases worldwide and has garnered increasing attention in recent decades. NAFLD is characterized by a wide range of liver changes, from simple steatosis to nonalcoholic steatohepatitis, cirrhosis, and hepatocellular carcinoma. The blurred pathogenesis of NAFLD is very complicated and involves lipid accumulation, insulin resistance, inflammation, and fibrogenesis. NAFLD is closely associated with complications such as obesity, diabetes, steatohepatitis, and liver fibrosis. During the progression of NAFLD, reactive oxygen species (ROS) are activated and induce oxidative stress. Recent attempts at establishing effective NAFLD therapy have identified potential micronutrient antioxidants that may reduce the accumulation of ROS and finally ameliorate the disease. In this review, we present the molecular mechanisms involved in the pathogenesis of NAFLD and introduce some dietary antioxidants that may be used to prevent or cure NAFLD, such as vitamin D, E, and astaxanthin.
Collapse
Affiliation(s)
- Guanliang Chen
- Department of Cell Metabolism and Nutrition, Brain/Liver Interface Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan.
| | - Yinhua Ni
- Department of Cell Metabolism and Nutrition, Brain/Liver Interface Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan.
| | - Naoto Nagata
- Department of Cell Metabolism and Nutrition, Brain/Liver Interface Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan.
| | - Liang Xu
- Department of Cell Metabolism and Nutrition, Brain/Liver Interface Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan.
| | - Tsuguhito Ota
- Department of Cell Metabolism and Nutrition, Brain/Liver Interface Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan.
| |
Collapse
|
50
|
Novel Action of Carotenoids on Non-Alcoholic Fatty Liver Disease: Macrophage Polarization and Liver Homeostasis. Nutrients 2016; 8:nu8070391. [PMID: 27347998 PMCID: PMC4963867 DOI: 10.3390/nu8070391] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/15/2016] [Accepted: 06/22/2016] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease. It is characterized by a wide spectrum of hepatic changes, which may progress to non-alcoholic steatohepatitis (NASH) and cirrhosis. NAFLD is considered a hepatic manifestation of metabolic syndrome; however, mechanisms underlying the onset and progression of NAFLD are still unclear. Resident and recruited macrophages are key players in the homeostatic function of the liver and in the progression of NAFLD to NASH. Progress has been made in understanding the molecular mechanisms underlying the polarized activation of macrophages. New NAFLD therapies will likely involve modification of macrophage polarization by restraining M1 activation or driving M2 activation. Carotenoids are potent antioxidants and anti-inflammatory micronutrients that have been used to prevent and treat NAFLD. In addition to their antioxidative action, carotenoids can regulate macrophage polarization and thereby halt the progression of NASH. In this review, we summarize the molecular mechanisms of macrophage polarization and the function of liver macrophages/Kupffer cells in NAFLD. From our review, we propose that dietary carotenoids, such as β-cryptoxanthin and astaxanthin, be used to prevent or treat NAFLD through the regulation of macrophage polarization and liver homeostasis.
Collapse
|