1
|
Liu Z, Hou J, Tian M, Zhang Y, Huang D, Zhao N, Ma Y, Cui S. Hypoxia ameliorates high-fat-diet-induced hepatic lipid accumulation by modulating the HIF2α/PP4C signaling. Cell Signal 2025; 131:111751. [PMID: 40112904 DOI: 10.1016/j.cellsig.2025.111751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/03/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Hepatic lipid accumulation is a hallmark of metabolically associated fatty liver disease (MAFLD), which contributes to the progression of cirrhosis and even hepatoma. However, the underlying mechanisms remain poorly understood. Protein phosphatase 4C (PP4C) is an important enzyme that exists widely in the body and participates in cell metabolism. Hypoxia can affect the development of metabolic diseases. In this study, we investigated the role of PP4C in hepatic lipid metabolism under hypoxia in vivo and in vitro. Hypoxia-inducible factor 2α (HIF2α), PP4C, phosphorylated AU-rich element RNA-binding factor 1(pAUF1), acetyl-CoA carboxylase 1 (ACC1), and carnitine palmitoyl transferase-1 (CPT1) were analyzed via western blotting and immunofluorescence. The mechanism by which PP4C affects hepatic lipid accumulation under hypoxia was evaluated in stable transfected cell lines. Compared with those in the 2200 m HFD group, body weight, triglyceride (TG), total cholesterol (TC), amino alanine transferase (ALT), aspartate transaminase (AST), and lipid accumulation were lower in the 4500 m HFD group (P < 0.05). Compared with those in the 4500 m ND group, ACC1 and PP4C levels were lower than in the 4500 m HFD group, but HIF2α, pAUF1, and CPT1 levels were greater (P < 0.05). Knockdown of HIF2α prevented the hypoxia-induced reduction of PP4C, confirming the regulatory role of the HIF2α-PP4C axis in hepatic lipid metabolism. PP4C could affect the phosphorylation and expression localization of AU-rich element RNA-binding factor 1 (AUF1). PP4C enhanced lipid accumulation by reducing pAUF1, while the knockdown of PP4C had the opposite effect; pAUF1 had no change. Compared with those in the control group, ACC1 levels were decreased and CPT1 levels were increased in the AUF1 overexpression group, whereas ACC1 and CPT1 levels were not altered in the AUF1 knockdown group (P < 0.05). In conclusion, hypoxia might improve lipid accumulation by downregulating PP4C via HIF2a. PP4C is involved in hepatic lipid metabolism by regulating AUF1 phosphorylation under different oxygen concentrations. PP4C might be a promising target for treating hepatic lipid accumulation.
Collapse
Affiliation(s)
- Zhe Liu
- Research Center for High Altitude Medicine, Qinghai University, Xining 810000, China; Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Xining 810000, China; Department of Gynecology, Affiliated Hospital of Qinghai University, Xining 810000, China
| | - Jing Hou
- Central Laboratory/Research Key Laboratory for Echinococcosis, Affiliated Hospital of Qinghai University, Xining 810000, China
| | - MeiYuan Tian
- Central Laboratory/Research Key Laboratory for Echinococcosis, Affiliated Hospital of Qinghai University, Xining 810000, China
| | - YaoGang Zhang
- Central Laboratory/Research Key Laboratory for Echinococcosis, Affiliated Hospital of Qinghai University, Xining 810000, China
| | - DengLiang Huang
- Central Laboratory/Research Key Laboratory for Echinococcosis, Affiliated Hospital of Qinghai University, Xining 810000, China
| | - Na Zhao
- Graduate School of Qinghai University, Qinghai University, Xining 810000, China
| | - Yanyan Ma
- Central Laboratory/Research Key Laboratory for Echinococcosis, Affiliated Hospital of Qinghai University, Xining 810000, China; Department of Scientific Research Office, Affiliated Hospital of Qinghai University, Xining 810000, China.
| | - Sen Cui
- Department of Hematology, Affiliated Hospital of Qinghai University, Xining 810000, China.
| |
Collapse
|
2
|
Kudo J, Hirono H, Ohkoshi S. Low-frequency, mild-gradient chronic intermittent hypoxia still induces liver fibrogenesis in mice on a high-fat diet. Biochem Biophys Res Commun 2025; 761:151744. [PMID: 40184789 DOI: 10.1016/j.bbrc.2025.151744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/03/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
The fibrogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD) may progress when complicated by obstructive sleep apnea (OSA). Studies using animal models have shown that high-frequency intermittent hypoxia (IH) exposure, which resembles human OSA, accelerates liver fibrosis in fatty liver. This study highlights that low-frequency, mild-gradient intermittent hypoxia (IH) can exacerbate fibrogenesis in fatty liver disease, even without significantly raising markers of liver injury or insulin resistance. Using a mice model on a high-fat diet (HFD), we found that while routine liver tests (e.g., ALT, AST) and cholesterol levels remained comparable between HFD mice exposed to room air (RA) versus those exposed to chronic intermittent hypoxia (CIH), indicators of liver fibrosis and oxidative stress were elevated in the latter group. This suggests that even low-frequency, mild-gradient IH can increase oxidative stress and fibrotic activity within the liver, primarily through the upregulation of specific markers like ICAM-1 and osteopontin (OPN), which may play a role CIH-induced liver inflammation and fibrosis in fatty liver. In conclusion. Our study further notes that these hypoxia-related changes occurred without significantly worsening systemic insulin resistance, focusing attention on localized liver impacts rather than global metabolic disruptions. The findings underscore the potential role of oxidative stress and specific cytokines in the progression of liver fibrosis in MASLD, especially when complicated by conditions that introduce IH.
Collapse
Affiliation(s)
- Junpei Kudo
- Clinical Examination, Graduate School of Life Dentistry at Niigata, The Nippon Dental University, Niigata, Japan
| | - Haruka Hirono
- Clinical Examination, Graduate School of Life Dentistry at Niigata, The Nippon Dental University, Niigata, Japan
| | - Shogo Ohkoshi
- Clinical Examination, Graduate School of Life Dentistry at Niigata, The Nippon Dental University, Niigata, Japan.
| |
Collapse
|
3
|
Caviglia GP, Casalone E, Rosso C, Aneli S, Allione A, Carli F, Grange C, Armandi A, Catalano C, Birolo G, Foglia B, Ribaldone DG, Gastaldelli A, Matullo G, Bugianesi E. Extracellular Vesicles miRNome Profiling Reveals miRNAs Engagement in Dysfunctional Lipid Metabolism, Chronic Inflammation and Liver Damage in Subjects With Metabolic Dysfunction-Associated Steatotic Liver Disease. Aliment Pharmacol Ther 2025. [PMID: 40208030 DOI: 10.1111/apt.70150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/03/2024] [Accepted: 04/04/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND AND AIMS MicroRNAs (miRNAs) are short non-coding oligonucleotides involved in the post-transcriptional regulation of gene expression. We investigated the association between the miRNome profile of circulating extracellular vesicles (EVs) and metabolic derangements, circulating and hepatic pro-inflammatory cytokines, and liver damage across the histological spectrum of metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS EV miRNAs expression was determined by NGS (NextSeq550, Illumina Inc) in 228 biopsy-proven MASLD patients. In vivo metabolic studies were performed in a subgroup of 54 patients by tracer infusion ([6,6-2H2]glucose and [2H5]glycerol) to assess glucose and lipid fluxes and insulin resistance (IR) in the adipose tissue. RESULTS Seven miRNAs (miR-27b-3p, miR-30a-5p, miR-122-5p, miR-375-3p, miR-103a-3p, let-7d-5p, and let-7f-5p) were differentially expressed according to the diagnosis of steatohepatitis and the presence of significant fibrosis (F ≥ 2), thus marking subjects with 'at-risk MASH'. In the metabolic studies, the above-reported miRNAs had the strongest associations with lipid metabolism: miR-122-5p and miR-375-3p levels directly correlated with circulating free fatty acids (FFAs) and adipose tissue (AT)-IR, while let-7d-5p and let-7f-5p inversely correlated with lipolysis, FFAs, and progressively decreased according to AT-IR severity. In addition, let-7d-5p and let-7f-5p inversely correlated with the circulating and hepatic expression of pro-inflammatory cytokines, which increased by increasing degrees of AT-IR. CONCLUSIONS Our results suggest an intertwined connection between miR-122-5p, miR-375-3p, and the let-7 family in modulating lipid derangements and inflammatory pathways in patients with 'at-risk MASH', paving the basis for further studies aiming at investigating their potential therapeutic value.
Collapse
Affiliation(s)
- Gian Paolo Caviglia
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Elisabetta Casalone
- Unit of Genomic, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Chiara Rosso
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Serena Aneli
- Unit of Genomic, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alessandra Allione
- Unit of Genomic, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Fabrizia Carli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, Pisa, Italy
| | - Cristina Grange
- Division of Internal Medicine, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Angelo Armandi
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Turin, Italy
- Metabolic Liver Disease Research Program, I. Department of Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Chiara Catalano
- Unit of Genomic, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giovanni Birolo
- Unit of Genomic, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Beatrice Foglia
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Davide Giuseppe Ribaldone
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, Pisa, Italy
| | - Giuseppe Matullo
- Unit of Genomic, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Elisabetta Bugianesi
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
4
|
Kong F, Lei L, Cai L, Li J, Zhao C, Liu M, Qi D, Gao J, Li E, Gao W, Du X, Song Y, Liu G, Li X. Hypoxia-inducible factor 2α mediates nonesterified fatty acids and hypoxia-induced lipid accumulation in bovine hepatocytes. J Dairy Sci 2025; 108:4062-4078. [PMID: 39890076 DOI: 10.3168/jds.2024-25839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/23/2024] [Indexed: 02/03/2025]
Abstract
Ketosis is a metabolic disorder frequently occurring in the perinatal period, characterized by elevated circulating concentrations of nonesterified fatty acids (NEFA) due to negative energy balance, resulting in fatty liver in dairy cows. However, the mechanism of hepatic steatosis induced by high concentrations of NEFA in ketosis remains unclear. Hypoxia-inducible factor 2α (HIF-2α), which mediates adaptation to hypoxic stress, plays a critical role in regulating lipid metabolism. In this study, we investigate whether HIF-2α is involved in NEFA-driven hepatic lipid accumulation in dairy cows with ketosis. Liver and blood samples were collected from 10 healthy cows (blood BHB concentration <1.2 mM) and 10 ketotic cows (blood BHB concentration >3.0 mM with clinical symptoms) with similar lactation numbers (median = 3, range = 2-4) at 3 to 9 DIM (median = 6). In cows with ketosis, serum concentrations of NEFA and BHB were greater, but serum concentrations of glucose were lower. Moreover, hepatic triglyceride content increased significantly. In the liver of ketotic cows, which was accompanied by upregulated HIF-2α expression. To determine the potential association among hypoxia, HIF-2α, and the formation of hepatocellular steatosis in vitro, we isolated hepatocytes from healthy calves for the following experiments. First, hepatocytes were treated with 0, 0.6, 1.2, or 2.4 mM NEFA (52.7 mM stock NEFA solution was diluted in RPMI-1640 basic medium supplemented with 2% fatty acid-free BSA to achieve the specified concentrations) for 18 h, showing that HIF-2α expression and cellular hypoxia occurred in a dose-dependent manner. Next, hepatocytes were infected with HIF-2α (encoded by EPAS1) small interfering RNA (Si-HIF-2α) for 48 h and then treated with 1.2 mM NEFA for 18 h. Results indicated that silencing HIF-2α decreased NEFA-induced lipid accumulation in bovine hepatocytes. Subsequently, hepatocytes treated with or without NEFA were placed in an AnaeroPack System, mimicking a hypoxic condition, for 0, 12, 18, or 24 h. Results showed that hypoxia could induce and further exacerbate lipid accumulation in bovine hepatocytes. Meanwhile, normal or NEFA-treated hepatocytes were cocultured with or without PT2385, a specific HIF-2α inhibitor, showing that hypoxia promoted steatosis through HIF-2α. Activating transcription factor 4 (ATF4) is an endoplasmic reticulum (ER) stress and hypoxia-inducible transcription factor. Here, bovine hepatocytes were treated with NEFA or hypoxia following transfecting ATF4 small interfering RNA, which demonstrated that ATF4 knockdown alleviated the extent of lipid accumulation in bovine hepatocytes. In addition, we found that ATF4 expression was correlated with HIF-2α levels in both liver tissue and cultured hepatocyte models. Moreover, overexpression of ATF4 weakened the beneficial effects of HIF-2α inhibition. Overall, these data suggest that NEFA-induced hepatic hypoxia significantly contributes to the progression of hepatic steatosis which in turn, intensifies hypoxia and leads to a self-perpetuating cycle of reciprocal causation, further exacerbating hepatic lipid deposition. Additionally, accumulated HIF-2α plays a critical role in this complex-origin steatosis, potentially through ATF4.
Collapse
Affiliation(s)
- Fanrong Kong
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Lin Lei
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Lin Cai
- College of Food and Biology of Changchun Polytechnic, Changchun 130062, China
| | - Jinxia Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Chenchen Zhao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Menglin Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Dandan Qi
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jie Gao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Enzhu Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wenwen Gao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiliang Du
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuxiang Song
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Guowen Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xinwei Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
5
|
Jiang M, Huang J, Guo X, Fu W, Peng L, Wang Y, Liu W, Liu J, Zhou L, Xiao Y. HIF-3α/PPAR-γ Regulates Hypoxia Tolerance by Altering Glycolysis and Lipid Synthesis in Blunt Snout Bream ( Megalobrama amblycephala). Int J Mol Sci 2025; 26:2613. [PMID: 40141255 PMCID: PMC11942064 DOI: 10.3390/ijms26062613] [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: 01/24/2025] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Hypoxic stress causes cell damage and serious diseases in organisms, especially in aquatic animals. It is important to elucidate the changes in metabolic function caused by hypoxia and the mechanisms underlying these changes. This study focuses on the low oxygen tolerance feature of a new blunt snout bream strain (GBSBF1). Our data show that GBSBF1 has a different lipid and carbohydrate metabolism pattern than wild-type bream, with altering glycolysis and lipid synthesis. In GBSBF1, the expression levels of phd2 and vhl genes are significantly decreased, while the activation of HIF-3α protein is observed to have risen significantly. The results indicate that enhanced HIF-3α can positively regulate gpd1ab and gpam through PPAR-γ, which increases glucose metabolism and reduces lipolysis of GBSBF1. This research is beneficial for creating new aquaculture strains with low oxygen tolerance traits.
Collapse
Affiliation(s)
- Minggui Jiang
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Jing Huang
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
| | - Xing Guo
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
| | - Wen Fu
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Liangyue Peng
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Yang Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Y.W.); (L.Z.)
| | - Wenbin Liu
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Jinhui Liu
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Li Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Y.W.); (L.Z.)
| | - Yamei Xiao
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| |
Collapse
|
6
|
Mooli RGR, Mukhi D, Watt M, Nagati V, Reed SM, Gandhi NK, Oertel M, Ramakrishnan SK. Hypoxia-Inducible Factor-2α Promotes Liver Fibrosis by Inducing Hepatocellular Death. Int J Mol Sci 2024; 25:13114. [PMID: 39684823 DOI: 10.3390/ijms252313114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
The activation of hypoxia-inducible factors (HIF)-1α and 2α in the liver is closely linked to the progression of fatty liver diseases. Prior studies indicated that disrupting hepatocyte HIF-2α attenuates diet-induced hepatic steatosis, subsequently decreasing fibrosis. However, the direct role of hepatocyte HIF-2α in liver fibrosis has not been addressed. Hepatic HIF-2α expression was examined in mouse model of carbon tetrachloride (CCl4)-induced liver fibrosis. Conditional hepatocyte Hif-2α knockout mice were employed to investigate the role of hepatocyte HIF-2α in fibrosis. Markers of apoptosis, proliferation, inflammation, and fibrosis were assessed through biochemical, molecular, and histological analyses. We found an induction of HIF-2α in CCL4-injected liver injury and fibrosis mouse models. Hepatocyte-specific deletion of HIF-2α attenuated stellate cell activation and fibrosis, with no significant difference in inflammation. Disrupting hepatocyte HIF-2α led to reduced injury-mediated hepatocellular apoptosis. Surviving hepatocytes exhibited hypertrophy, which was strongly associated with the activation of c-JUN signaling. Our study demonstrates a direct role of hepatocyte HIF-2α in liver fibrosis by promoting hepatocyte apoptosis. The reduction in apoptosis and induction of hepatocyte hypertrophy following HIF-2α disruption is closely linked to enhanced c-JUN signaling, a survival mechanism in response to liver injury. These findings highlight HIF-2α as a potential therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Raja Gopal Reddy Mooli
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Dhanunjay Mukhi
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Mikayla Watt
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Veerababu Nagati
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Sara M Reed
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Nikita K Gandhi
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Michael Oertel
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Sadeesh K Ramakrishnan
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
7
|
Qi F, Li T, Deng Q, Fan A. The impact of aerobic and anaerobic exercise interventions on the management and outcomes of non-alcoholic fatty liver disease. Physiol Res 2024; 73:671-686. [PMID: 39530904 PMCID: PMC11629946 DOI: 10.33549/physiolres.935244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 06/25/2024] [Indexed: 12/13/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a metabolic disorder that includes non-alcoholic hepatic steatosis without or with moderate inflammation and non-alcoholic steatohepatitis (NASH), characterized by necroinflammation and a more rapid progression of fibrosis. It is the primary pathological basis for hepatocellular carcinoma. With its prevalence escalating annually, NAFLD has emerged as a global health epidemic, presenting a significant hazard to public health worldwide. Existing studies have shown that physical activity and exercise training have a positive effect on NAFLD. However, the extent to which exercise improves NAFLD depends on the type, intensity, and duration. Therefore, the type of exercise that has the best effect on improving NAFLD remains to be explored. To date, the most valuable discussions involve aerobic and anaerobic exercise. Exercise intervenes in the pathological process of NAFLD by regulating physiological changes in cells through multiple signaling pathways. The review aims to summarize the signaling pathways affected by two different exercise types associated with the onset and progression of NAFLD. It provides a new basis for improving and managing NAFLD in clinical practice.
Collapse
Affiliation(s)
- F Qi
- Chongqing College of International Business and Economics, Southwest University, Chongqing, China, College of Physical Education, Southwest University, Chongqing, China.
| | | | | | | |
Collapse
|
8
|
Villano G, Pontisso P. Protease activated receptor 2 as a novel druggable target for the treatment of metabolic dysfunction-associated fatty liver disease and cancer. Front Immunol 2024; 15:1397441. [PMID: 39464875 PMCID: PMC11502361 DOI: 10.3389/fimmu.2024.1397441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 09/23/2024] [Indexed: 10/29/2024] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is spreading worldwide, largely due to unhealthy lifestyles that contribute to the rise in diabetes, metabolic syndrome, and obesity. In this situation, the progression of injury to metabolic steatohepatitis can evolve to cirrhosis and, eventually, to hepatocellular carcinoma (HCC). It is well known that serine protease enzymes with different functions in cellular homeostasis act as signaling molecules that regulate liver inflammation by activating the protease-activated receptors (PARs) family members, expressed on the cellular plasma membrane. Among them, PAR2 plays a central role in the activation of signaling pathways in response to changes in the extracellular microenvironment. Experimental data have provided evidence that PAR2 is involved not only in inflammatory response but also in insulin resistance, lipid metabolism, and cancer. The major aims of this narrative review are addressed to assess PAR2 involvement in inflammation, metabolism, and liver disease progression and to explore possible therapeutic strategies, based on PAR2 inhibition, in order to prevent its biological effects in the context of MAFLD and cancer.
Collapse
Affiliation(s)
- Gianmarco Villano
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova, Italy
| | | |
Collapse
|
9
|
Jeelani I, Moon JS, da Cunha FF, Nasamran CA, Jeon S, Zhang X, Bandyopadhyay GK, Dobaczewska K, Mikulski Z, Hosseini M, Liu X, Kisseleva T, Brenner D, Singh S, Loomba R, Kim M, Lee YS. HIF-2α drives hepatic Kupffer cell death and proinflammatory recruited macrophage activation in nonalcoholic steatohepatitis. Sci Transl Med 2024; 16:eadi0284. [PMID: 39259813 PMCID: PMC11665927 DOI: 10.1126/scitranslmed.adi0284] [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: 03/29/2023] [Revised: 01/12/2024] [Accepted: 08/16/2024] [Indexed: 09/13/2024]
Abstract
Proinflammatory hepatic macrophage activation plays a key role in the development of nonalcoholic steatohepatitis (NASH). This involves increased embryonic hepatic Kupffer cell (KC) death, facilitating the replacement of KCs with bone marrow-derived recruited hepatic macrophages (RHMs) that highly express proinflammatory genes. Moreover, phago/efferocytic activity of KCs is diminished in NASH, enhancing liver inflammation. However, the molecular mechanisms underlying these changes in KCs are not known. Here, we show that hypoxia-inducible factor 2α (HIF-2α) mediates NASH-associated decreased KC growth and efferocytosis by enhancing lysosomal stress. At the molecular level, HIF-2α stimulated mammalian target of rapamycin (mTOR)- and extracellular signal-regulated kinase-dependent inhibitory transcription factor EB (TFEB) phosphorylation, leading to decreased lysosomal and phagocytic gene expression. With increased metabolic stress and phago/efferocytic burden in NASH, these changes were sufficient to increase lysosomal stress, causing decreased efferocytosis and lysosomal cell death. Of interest, HIF-2α-dependent TFEB regulation only occurred in KCs but not RHMs. Instead, in RHMs, HIF-2α promoted mitochondrial reactive oxygen species production and proinflammatory activation by increasing ANT2 expression and mitochondrial permeability transition. Consequently, myeloid lineage-specific or KC-specific HIF-2α depletion or the inhibition of mTOR-dependent TFEB inhibition using antisense oligonucleotide treatment protected against the development of NASH in mice. Moreover, treatment with an HIF-2α-specific inhibitor reduced inflammatory and fibrogenic gene expression in human liver spheroids cultured under a NASH-like condition. Together, our results suggest that macrophage subtype-specific effects of HIF-2α collectively contribute to the proinflammatory activation of liver macrophages, leading to the development of NASH.
Collapse
Affiliation(s)
- Ishtiaq Jeelani
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California, 92093, USA
| | - Jae-Su Moon
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California, 92093, USA
| | - Flavia Franco da Cunha
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California, 92093, USA
| | - Chanond A. Nasamran
- Center for Computational Biology & Bioinformatics, Department of Medicine, University of California San Diego, La Jolla, California, 92093, USA
| | - Seokhyun Jeon
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California, 92093, USA
| | - Xinhang Zhang
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California, 92093, USA
| | - Gautam K. Bandyopadhyay
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California, 92093, USA
| | - Katarzyna Dobaczewska
- Microscopy and Histology Core Facility, La Jolla Institute for Immunology, La Jolla, California, 92037, USA
| | - Zbigniew Mikulski
- Microscopy and Histology Core Facility, La Jolla Institute for Immunology, La Jolla, California, 92037, USA
| | - Mojgan Hosseini
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, California, 92093, USA
| | - Xiao Liu
- Department of Surgery, University of California San Diego, La Jolla, California, 92093, USA
| | - Tatiana Kisseleva
- Department of Surgery, University of California San Diego, La Jolla, California, 92093, USA
| | - David Brenner
- Department of Medicine, University of California San Diego, La Jolla, California, 92093, USA
| | - Seema Singh
- Division of Gastroenterology, University of California San Diego, La Jolla, California, 92093, USA
| | - Rohit Loomba
- Division of Gastroenterology, University of California San Diego, La Jolla, California, 92093, USA
- Division of Epidemiology Department of Family Medicine and Public Health, University of California, San Diego, La Jolla, California, 92093, USA
- NAFLD Research Center University of California, San Diego, La Jolla, California, 92093, USA
| | - Minkyu Kim
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, Texas, 78229, USA
| | - Yun Sok Lee
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California, 92093, USA
| |
Collapse
|
10
|
Cagnin S, Pontisso P, Martini A. SerpinB3: A Multifaceted Player in Health and Disease-Review and Future Perspectives. Cancers (Basel) 2024; 16:2579. [PMID: 39061218 PMCID: PMC11274807 DOI: 10.3390/cancers16142579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/10/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
SerpinB3, a member of the serine-protease inhibitor family, has emerged as a crucial player in various physiological and pathological processes. Initially identified as an oncogenic factor in squamous cell carcinomas, SerpinB3's intricate involvement extends from fibrosis progression and cancer to cell protection in acute oxidative stress conditions. This review explores the multifaceted roles of SerpinB3, focusing on its implications in fibrosis, metabolic syndrome, carcinogenesis and immune system impairment. Furthermore, its involvement in tissue protection from oxidative stress and wound healing underscores its potential as diagnostic and therapeutic tool. Recent studies have described the therapeutic potential of targeting SerpinB3 through its upstream regulators, offering novel strategies for cancer treatment development. Overall, this review underscores the importance of further research to fully elucidate the mechanisms of action of SerpinB3 and to exploit its therapeutic potential across various medical conditions.
Collapse
Affiliation(s)
| | - Patrizia Pontisso
- Department of Medicine, University of Padova, 35123 Padova, Italy; (S.C.); (A.M.)
| | | |
Collapse
|
11
|
Atallah E, Trehiou S, Alquier-Bacquie V, Lasserre F, Arroyo J, Molette C, Remignon H. Development of hepatic steatosis in male and female mule ducks after respective force-feeding programs. Front Physiol 2024; 15:1392968. [PMID: 38974520 PMCID: PMC11224645 DOI: 10.3389/fphys.2024.1392968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/24/2024] [Indexed: 07/09/2024] Open
Abstract
Male and female mule ducks were subjected to a force-feeding diet to induce liver steatosis as it is generally done only with male ducks for the production of foie gras. The different biochemical measurements indicated that the course of hepatic steatosis development was present in both sexes and associated with a huge increase in liver weight mainly due to the synthesis and accumulation of lipids in hepatocytes. In livers of male and female ducks, this lipid accumulation was associated with oxidative stress and hypoxia. However, certain specific modifications (kinetics of lipid droplet development and hepatic inflammation) indicate that female ducks may tolerate force-feeding less well, at least at the hepatic level. This is in contradiction with what is generally reported concerning hepatic steatosis induced by dietary disturbances in mammals but could be explained by the very specific conditions imposed by force-feeding. Despite this, force-feeding female ducks seems entirely feasible, provided that the final quality of the product is as good as that of the male ducks, which will remain to be demonstrated in future studies.
Collapse
Affiliation(s)
- Elham Atallah
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, UPS, Université de Toulouse, Toulouse, France
| | - Sabrina Trehiou
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, UPS, Université de Toulouse, Toulouse, France
| | - Valérie Alquier-Bacquie
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, UPS, Université de Toulouse, Toulouse, France
| | - Frédéric Lasserre
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, UPS, Université de Toulouse, Toulouse, France
| | | | | | - Hervé Remignon
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, UPS, Université de Toulouse, Toulouse, France
- INP-ENSAT, Université de Toulouse, Castanet-Tolosan, France
| |
Collapse
|
12
|
Xia J, Chen H, Wang X, Chen W, Lin J, Xu F, Nie Q, Ye C, Zhong B, Zhao M, Yun C, Zeng G, Mao Y, Wen Y, Zhang X, Yan S, Wang X, Sun L, Liu F, Zhong C, Xia P, Jiang C, Rao H, Pang Y. Sphingosine d18:1 promotes nonalcoholic steatohepatitis by inhibiting macrophage HIF-2α. Nat Commun 2024; 15:4755. [PMID: 38834568 PMCID: PMC11150497 DOI: 10.1038/s41467-024-48954-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 05/17/2024] [Indexed: 06/06/2024] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a severe type of the non-alcoholic fatty liver disease (NAFLD). NASH is a growing global health concern due to its increasing morbidity, lack of well-defined biomarkers and lack of clinically effective treatments. Using metabolomic analysis, the most significantly changed active lipid sphingosine d18:1 [So(d18:1)] is selected from NASH patients. So(d18:1) inhibits macrophage HIF-2α as a direct inhibitor and promotes the inflammatory factors secretion. Male macrophage-specific HIF-2α knockout and overexpression mice verified the protective effect of HIF-2α on NASH progression. Importantly, the HIF-2α stabilizer FG-4592 alleviates liver inflammation and fibrosis in NASH, which indicated that macrophage HIF-2α is a potential drug target for NASH treatment. Overall, this study confirms that So(d18:1) promotes NASH and clarifies that So(d18:1) inhibits the transcriptional activity of HIF-2α in liver macrophages by suppressing the interaction of HIF-2α with ARNT, suggesting that macrophage HIF-2α may be a potential target for the treatment of NASH.
Collapse
Affiliation(s)
- Jialin Xia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking University, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Hong Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking University, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Xiaoxiao Wang
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Weixuan Chen
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Jun Lin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking University, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Feng Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking University, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Qixing Nie
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking University, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chuan Ye
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking University, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Bitao Zhong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking University, Beijing, China
| | - Min Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Chuyu Yun
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Guangyi Zeng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking University, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Yuejian Mao
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Yongping Wen
- Mengniu Institute of Nutrition Science, Shanghai, China
| | - Xuguang Zhang
- Mengniu Institute of Nutrition Science, Shanghai, China
- Shanghai Institute of Nutrition and Health, The Chinese Academy of Sciences, Shanghai, China
| | - Sen Yan
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Xuemei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking University, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Lulu Sun
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China
| | - Feng Liu
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Chao Zhong
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing, China
| | - Pengyan Xia
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking University, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing, China
| | - Huiying Rao
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China.
| | - Yanli Pang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking University, Beijing, China.
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China.
| |
Collapse
|
13
|
Deng Y, Hu M, Huang S, Fu N. Molecular mechanism and therapeutic significance of essential amino acids in metabolically associated fatty liver disease. J Nutr Biochem 2024; 126:109581. [PMID: 38219809 DOI: 10.1016/j.jnutbio.2024.109581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/01/2024] [Accepted: 01/06/2024] [Indexed: 01/16/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD), also known as metabolically associated fatty liver disease (MAFLD), is a systemic metabolic disease characterized by lipid accumulation in the liver, lipid toxicity, insulin resistance, intestinal dysbiosis, and inflammation that can progress from simple steatosis to nonalcoholic steatohepatitis (NASH) and even cirrhosis or cancer. It is the most prevalent illness threatening world health. Currently, there are almost no approved drug interventions for MAFLD, mainly dietary changes and exercise to control weight and regulate metabolic disorders. Meanwhile, the metabolic pathway involved in amino acid metabolism also influences the onset and development of MAFLD in the body, and most amino acid metabolism takes place in the liver. Essential amino acids are those amino acids that must be supplemented from outside the diet and that cannot be synthesized in the body or cannot be synthesized at a rate sufficient to meet the body's needs, including leucine, isoleucine, valine (collectively known as branched-chain amino acids), tryptophan, phenylalanine (which are aromatic amino acids), histidine, methionine, threonine and lysine. The metabolic balance of the body is closely linked to these essential amino acids, and essential amino acids are closely linked to the pathophysiological process of MAFLD. In this paper, we will focus on the metabolism of essential amino acids in the body and further explore the therapeutic strategies for MAFLD based on the studies conducted in recent years.
Collapse
Affiliation(s)
- Yuting Deng
- The Affiliated Nanhua Hospital, Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Mengsi Hu
- The Affiliated Nanhua Hospital, Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Shufang Huang
- The Affiliated Nanhua Hospital, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China.
| | - Nian Fu
- The Affiliated Nanhua Hospital, Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China; The Affiliated Nanhua Hospital, Institute of Clinical Research, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China.
| |
Collapse
|
14
|
Villano G, Novo E, Turato C, Quarta S, Ruvoletto M, Biasiolo A, Protopapa F, Chinellato M, Martini A, Trevellin E, Granzotto M, Cannito S, Cendron L, De Siervi S, Guido M, Parola M, Vettor R, Pontisso P. The protease activated receptor 2 - CCAAT/enhancer-binding protein beta - SerpinB3 axis inhibition as a novel strategy for the treatment of non-alcoholic steatohepatitis. Mol Metab 2024; 81:101889. [PMID: 38307387 PMCID: PMC10864841 DOI: 10.1016/j.molmet.2024.101889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/11/2024] [Accepted: 01/26/2024] [Indexed: 02/04/2024] Open
Abstract
OBJECTIVE The serine protease inhibitor SerpinB3 has been described as critical mediator of liver fibrosis and it has been recently proposed as an additional hepatokine involved in NASH development and insulin resistance. Protease Activated Receptor 2 has been identified as a novel regulator of hepatic metabolism. A targeted therapeutic strategy for NASH has been investigated, using 1-Piperidine Propionic Acid (1-PPA), since this compound has been recently proposed as both Protease Activated Receptor 2 and SerpinB3 inhibitor. METHODS The effect of SerpinB3 on inflammation and fibrosis genes was assessed in human macrophage and stellate cell lines. Transgenic mice, either overexpressing SerpinB3 or carrying Serpinb3 deletion and their relative wild type strains, were used in experimental NASH models. Subgroups of SerpinB3 transgenic mice and their controls were also injected with 1-PPA to assess the efficacy of this compound in NASH inhibition. RESULTS 1-PPA did not present significant cell and organ toxicity and was able to inhibit SerpinB3 and PAR2 in a dose-dependent manner. This effect was associated to a parallel reduction of the synthesis of the molecules induced by endogenous SerpinB3 or by its paracrine effects both in vitro and in vivo, leading to inhibition of lipid accumulation, inflammation and fibrosis in experimental NASH. At mechanistic level, the antiprotease activity of SerpinB3 was found essential for PAR2 activation, determining upregulation of the CCAAT Enhancer Binding Protein beta (C/EBP-β), another pivotal regulator of metabolism, inflammation and fibrosis, which in turn determined SerpinB3 synthesis. CONCLUSIONS 1-PPA treatment was able to inhibit the PAR2 - C/EBP-β - SerpinB3 axis and to protect from NASH development and progression, supporting the potential use of a similar approach for a targeted therapy of NASH.
Collapse
Affiliation(s)
- Gianmarco Villano
- Dept. of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Italy
| | - Erica Novo
- Dept. of Clinical and Biological Sciences, University of Torino, Italy
| | | | | | | | | | | | | | | | | | | | - Stefania Cannito
- Dept. of Clinical and Biological Sciences, University of Torino, Italy
| | | | | | - Maria Guido
- Dept. of Medicine, University of Padova, Italy
| | - Maurizio Parola
- Dept. of Clinical and Biological Sciences, University of Torino, Italy
| | | | | |
Collapse
|
15
|
Yan R, Cai H, Zhou X, Bao G, Bai Z, Ge RL. Hypoxia-inducible factor-2α promotes fibrosis in non-alcoholic fatty liver disease by enhancing glutamine catabolism and inhibiting yes-associated protein phosphorylation in hepatic stellate cells. Front Endocrinol (Lausanne) 2024; 15:1344971. [PMID: 38501098 PMCID: PMC10946064 DOI: 10.3389/fendo.2024.1344971] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/25/2024] [Indexed: 03/20/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has a high global prevalence and affects approximately one-third of adults, owing to high-fat dietary habits and a sedentary lifestyle. The role of hypoxia-inducible factor 2α (HIF-2α) in NAFLD progression remains unknown. This study aimed to investigate the effects of chronic hypoxia on NAFLD progression by examining the role of hypoxia-inducible factor 2α (HIF-2α) activation and that of hepatic stellate cell (HSC)-derived myofibroblasts through glutaminolysis. We hypothesised that hypoxia exacerbates NAFLD by promoting HIF-2α upregulation and inhibiting phosphorylated yes-associated protein (YAP), and that increasing YAP expression enhances HSC-derived myofibroblasts. We studied patients with NAFLD living at high altitudes, as well as animal models and cultured cells. The results revealed significant increases in HSC-derived myofibroblasts and collagen accumulation caused by HIF-2α and YAP upregulation, both in patients and in a mouse model for hypoxia and NAFLD. HIF-2α and HIF-2α-dependent YAP downregulation reduced HSC activation and myofibroblast levels in persistent chronic hypoxia. Furthermore, hypoxia-induced HIF-2α upregulation promoted YAP and inhibited YAP phosphorylation, leading to glutaminase 1 (GLS1), SLC38A1, α-SMA, and Collagen-1 overexpression. Additionally, hypoxia restored mitochondrial adenosine triphosphate production and reactive oxygen species (ROS) overproduction. Thus, chronic hypoxia-induced HIF-2α activation enhances fibrosis and NAFLD progression by restoring mitochondrial ROS production and glutaminase-1-induced glutaminolysis, which is mediated through the inhibition of YAP phosphorylation and increased YAP nuclear translocation. In summary, HIF-2α plays a pivotal role in NAFLD progression during chronic hypoxia.
Collapse
Affiliation(s)
- Ranran Yan
- Qinghai-Utah Joint Key Lab for High-altitude Medicine, Medical College of Qinghai University, Xining, China
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, China
- Key Laboratory of High-Altitude Medicine in Qinghai University, Ministry of Education, Xining, China
- Key Laboratory for Application of High-Altitude Medicine in Qinghai Province, Xining, China
| | - Hao Cai
- Oncology Department, The Fifth People’s Hospital of Qinghai Provincial, Xining, China
| | - Xiaofeng Zhou
- Affiliated Hospital of Qinghai University, Xining, China
| | - Guodan Bao
- Qinghai-Utah Joint Key Lab for High-altitude Medicine, Medical College of Qinghai University, Xining, China
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, China
- Key Laboratory of High-Altitude Medicine in Qinghai University, Ministry of Education, Xining, China
- Affiliated Hospital of Qinghai University, Xining, China
| | - Zhenzhong Bai
- Qinghai-Utah Joint Key Lab for High-altitude Medicine, Medical College of Qinghai University, Xining, China
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, China
- Key Laboratory of High-Altitude Medicine in Qinghai University, Ministry of Education, Xining, China
- Key Laboratory for Application of High-Altitude Medicine in Qinghai Province, Xining, China
| | - Ri-li Ge
- Qinghai-Utah Joint Key Lab for High-altitude Medicine, Medical College of Qinghai University, Xining, China
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, China
- Key Laboratory of High-Altitude Medicine in Qinghai University, Ministry of Education, Xining, China
- Key Laboratory for Application of High-Altitude Medicine in Qinghai Province, Xining, China
| |
Collapse
|
16
|
Foglia B, Sutti S, Cannito S, Rosso C, Maggiora M, Casalino A, Bocca C, Novo E, Protopapa F, Ramavath NN, Provera A, Gambella A, Bugianesi E, Tacke F, Albano E, Parola M. Histidine-rich glycoprotein in metabolic dysfunction-associated steatohepatitis-related disease progression and liver carcinogenesis. Front Immunol 2024; 15:1342404. [PMID: 38469298 PMCID: PMC10925642 DOI: 10.3389/fimmu.2024.1342404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/31/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD), previously non-alcoholic fatty liver disease (NAFLD), is a leading cause of chronic liver disease worldwide. In 20%-30% of MASLD patients, the disease progresses to metabolic dysfunction-associated steatohepatitis (MASH, previously NASH) which can lead to fibrosis/cirrhosis, liver failure as well as hepatocellular carcinoma (HCC). Here we investigated the role of histidine-rich glycoprotein (HRG), a plasma protein produced by hepatocytes, in MASLD/MASH progression and HCC development. METHODS The role of HRG was investigated by morphological, cellular, and molecular biology approaches in (a) HRG knock-out mice (HRG-/- mice) fed on a CDAA dietary protocol or a MASH related diethyl-nitrosamine/CDAA protocol of hepatocarcinogenesis, (b) THP1 monocytic cells treated with purified HRG, and (c) well-characterized cohorts of MASLD patients with or without HCC. RESULTS In non-neoplastic settings, murine and clinical data indicate that HRG increases significantly in parallel with disease progression. In particular, in MASLD/MASH patients, higher levels of HRG plasma levels were detected in subjects with extensive fibrosis/cirrhosis. When submitted to the pro-carcinogenic protocol, HRG-/- mice showed a significant decrease in the volume and number of HCC nodules in relation to decreased infiltration of macrophages producing pro-inflammatory mediators, including IL-1β, IL-6, IL-12, IL-10, and VEGF as well as impaired angiogenesis. The histopathological analysis (H-score) of MASH-related HCC indicate that the higher HRG positivity in peritumoral tissue significantly correlates with a lower overall patient survival and an increased recurrence. Moreover, a significant increase in HRG plasma levels was detected in cirrhotic (F4) patients and in patients carrying HCC vs. F0/F1 patients. CONCLUSION Murine and clinical data indicate that HRG plays a significant role in MASLD/MASH progression to HCC by supporting a specific population of tumor-associated macrophages with pro-inflammatory response and pro-angiogenetic capabilities which critically support cancer cell survival. Furthermore, our data suggest HRG as a possible prognostic predictor in HCC patients with MASLD/MASH-related HCCs.
Collapse
Affiliation(s)
- Beatrice Foglia
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Salvatore Sutti
- Department Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University Amedeo Avogadro of Eastern Piedmont, Novara, Italy
| | - Stefania Cannito
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Chiara Rosso
- Department Medical Sciences, University of Torino, and Division of Gastroenterology, San Giovanni Hospital, Torino, Italy
| | - Marina Maggiora
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Alice Casalino
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Claudia Bocca
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Erica Novo
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Francesca Protopapa
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Naresh Naik Ramavath
- Department of Pediatrics, School of Medicine, Washington University, St Louis, MO, United States
| | - Alessia Provera
- Department Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University Amedeo Avogadro of Eastern Piedmont, Novara, Italy
| | - Alessandro Gambella
- Department Medical Sciences, University of Torino, and Division of Gastroenterology, San Giovanni Hospital, Torino, Italy
| | - Elisabetta Bugianesi
- Department Medical Sciences, University of Torino, and Division of Gastroenterology, San Giovanni Hospital, Torino, Italy
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité-Universitatsmedizin Berlin, Berlin, Germany
| | - Emanuele Albano
- Department Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University Amedeo Avogadro of Eastern Piedmont, Novara, Italy
| | - Maurizio Parola
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| |
Collapse
|
17
|
Parola M, Pinzani M. Liver fibrosis in NAFLD/NASH: from pathophysiology towards diagnostic and therapeutic strategies. Mol Aspects Med 2024; 95:101231. [PMID: 38056058 DOI: 10.1016/j.mam.2023.101231] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023]
Abstract
Liver fibrosis, as an excess deposition of extracellular matrix (ECM) components, results from chronic liver injury as well as persistent activation of inflammatory response and of fibrogenesis. Liver fibrosis is a major determinant for chronic liver disease (CLD) progression and in the last two decades our understanding on the major molecular and cellular mechanisms underlying the fibrogenic progression of CLD has dramatically improved, boosting pre-clinical studies and clinical trials designed to find novel therapeutic approaches. From these studies several critical concepts have emerged, starting to reveal the complexity of the pro-fibrotic microenvironment which involves very complex, dynamic and interrelated interactions between different hepatic and extrahepatic cell populations. This review will offer first a recapitulation of established and novel pathophysiological basic principles and concepts by intentionally focus the attention on NAFLD/NASH, a metabolic-related form of CLD with a high impact on the general population and emerging as a leading cause of CLD worldwide. NAFLD/NASH-related pro-inflammatory and profibrogenic mechanisms will be analysed as well as novel information on cells, mediators and signalling pathways which have taken advantage from novel methodological approaches and techniques (single cell genomics, imaging mass cytometry, novel in vitro two- and three-dimensional models, etc.). We will next offer an overview on recent advancement in diagnostic and prognostic tools, including serum biomarkers and polygenic scores, to support the analysis of liver biopsies. Finally, this review will provide an analysis of current and emerging therapies for the treatment of NAFLD/NASH patients.
Collapse
Affiliation(s)
- Maurizio Parola
- Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Corso Raffaello 30, 10125, Torino, Italy.
| | - Massimo Pinzani
- UCL Institute for Liver and Digestive Health, Division of Medicine - Royal Free Hospital, London, NW32PF, United Kingdom.
| |
Collapse
|
18
|
Solanki S, Shah YM. Hypoxia-Induced Signaling in Gut and Liver Pathobiology. ANNUAL REVIEW OF PATHOLOGY 2024; 19:291-317. [PMID: 37832943 DOI: 10.1146/annurev-pathmechdis-051122-094743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
Oxygen (O2) is essential for cellular metabolism and biochemical reactions. When the demand for O2 exceeds the supply, hypoxia occurs. Hypoxia-inducible factors (HIFs) are essential to activate adaptive and survival responses following hypoxic stress. In the gut (intestines) and liver, the presence of oxygen gradients or physiologic hypoxia is necessary to maintain normal homeostasis. While physiologic hypoxia is beneficial and aids in normal functions, pathological hypoxia is harmful as it exacerbates inflammatory responses and tissue dysfunction and is a hallmark of many cancers. In this review, we discuss the role of gut and liver hypoxia-induced signaling, primarily focusing on HIFs, in the physiology and pathobiology of gut and liver diseases. Additionally, we examine the function of HIFs in various cell types during gut and liver diseases, beyond intestinal epithelial and hepatocyte HIFs. This review highlights the importance of understanding hypoxia-induced signaling in the pathogenesis of gut and liver diseases and emphasizes the potential of HIFs as therapeutic targets.
Collapse
Affiliation(s)
- Sumeet Solanki
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA;
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA;
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
19
|
Jia S, Li X, Du Q. Host insulin resistance caused by Porphyromonas gingivalis-review of recent progresses. Front Cell Infect Microbiol 2023; 13:1209381. [PMID: 37520442 PMCID: PMC10373507 DOI: 10.3389/fcimb.2023.1209381] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Porphyromonas gingivalis (P. gingivalis) is a Gram-negative oral anaerobic bacterium that plays a key role in the pathogenesis of periodontitis. P. gingivalis expresses a variety of virulence factors that disrupt innate and adaptive immunity, allowing P. gingivalis to survive and multiply in the host and destroy periodontal tissue. In addition to periodontal disease, P.gingivalis is also associated with systemic diseases, of which insulin resistance is an important pathological basis. P. gingivalis causes a systemic inflammatory response, disrupts insulin signaling pathways, induces pancreatic β-cell hypofunction and reduced numbers, and causes decreased insulin sensitivity leading to insulin resistance (IR). In this paper, we systematically review the studies on the mechanism of insulin resistance induced by P. gingivalis, discuss the association between P. gingivalis and systemic diseases based on insulin resistance, and finally propose relevant therapeutic approaches. Overall, through a systematic review of the mechanisms related to systemic diseases caused by P. gingivalis through insulin resistance, we hope to provide new insights for future basic research and clinical interventions for related systemic diseases.
Collapse
Affiliation(s)
- Shuxian Jia
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xiaobing Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Qin Du
- Department of Stomatology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
20
|
Quarta S, Cappon A, Turato C, Ruvoletto M, Cannito S, Villano G, Biasiolo A, Maggi M, Protopapa F, Bertazza L, Fasolato S, Parola M, Pontisso P. SerpinB3 Upregulates Low-Density Lipoprotein Receptor-Related Protein (LRP) Family Members, Leading to Wnt Signaling Activation and Increased Cell Survival and Invasiveness. BIOLOGY 2023; 12:771. [PMID: 37372056 DOI: 10.3390/biology12060771] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023]
Abstract
Abnormal activation of the Wnt-β-catenin signaling cascade is involved in tumor growth and dissemination. SerpinB3 has been shown to induce β-catenin, and both molecules are overexpressed in tumors, particularly in those with poor prognoses. The aim of this study was to evaluate the ability of SerpinB3 to modulate the Wnt pathway in liver cancer and in monocytic cells, the main type of inflammatory cells in the tumor microenvironment. The Wnt cascade, Wnt co-receptors, and low-density lipoprotein receptor-related protein (LRP) members were analyzed in different cell lines and human monocytes in the presence or absence of SerpinB3. The Wnt-β-catenin axis was also evaluated in liver tumors induced in mice with different extents of SeprinB3 expression. In monocytic cells, SerpinB3 induced a significant upregulation of Wnt-1/7, nuclear β-catenin, and c-Myc, which are associated with increased cell lifespan and proliferation. In liver tumors in mice, the expression of β-catenin was significantly correlated with the presence of SerpinB3. In hepatoma cells, Wnt co-receptors LRP-5/6 and LRP-1, implicated in cell survival and invasiveness, were upregulated by SerpinB3. The LRP pan-inhibitor RAP not only induced a decrease in LRP expression, but also a dose-dependent reduction in SerpinB3-induced invasiveness. In conclusion, SerpinB3 determines the activation of the Wnt canonical pathway and cell invasiveness through the upregulation of LRP family members.
Collapse
Affiliation(s)
- Santina Quarta
- Department of Medicine, University of Padova, 35128 Padua, Italy
| | - Andrea Cappon
- Department of Medicine, University of Padova, 35128 Padua, Italy
| | - Cristian Turato
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | | | - Stefania Cannito
- Department of Clinical and Biological Sciences, University of Torino, 10124 Turin, Italy
| | - Gianmarco Villano
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, 35128 Padua, Italy
| | | | - Maristella Maggi
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Francesca Protopapa
- Department of Clinical and Biological Sciences, University of Torino, 10124 Turin, Italy
| | - Loris Bertazza
- Department of Medicine, University of Padova, 35128 Padua, Italy
| | - Silvano Fasolato
- Department of Medicine, University of Padova, 35128 Padua, Italy
| | - Maurizio Parola
- Department of Clinical and Biological Sciences, University of Torino, 10124 Turin, Italy
| | | |
Collapse
|
21
|
Ye J, Chen J, Li Y, Sun L, Lu H. Hepatocyte-specific knockout of HIF-2α cannot alleviate carbon tetrachloride-induced liver fibrosis in mice. PeerJ 2023; 11:e15191. [PMID: 37033734 PMCID: PMC10078453 DOI: 10.7717/peerj.15191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/15/2023] [Indexed: 04/05/2023] Open
Abstract
Background
The effects of hypoxia inducible factor-2α (HIF-2α) deficiency on liver fibrosis have not been demonstrated in a fibrosis model induced by carbon tetrachloride (CCl4). We aimed to examine whether hepatocyte-specific HIF-2α deletion could ameliorate CCl4-induced liver fibrosis in mice.
Methods
Hepatocyte-specific HIF-2α knockout mice were created using an albumin promoter-driven Cre recombinase. HIF-2α knockout (KO) mice and floxed control wild-type (WT) mice were fed a normal diet (ND) and received either twice weekly intraperitoneal injections of CCl4 solution (CCl4 dissolved in olive oil) or the corresponding amount of olive oil for 8 weeks. The indicators of liver function, glucose and lipid metabolism, and liver histology were compared among the different groups.
Results
Hepatocyte-specific HIF-2α knockout had no effect on the growth, liver function, glucose or lipid metabolism in mice. CCl4-treated KO and WT mice had a similar pattern of injury and inflammatory cell infiltration in the liver. Quantification of Masson staining, α-smooth muscle actin (α-SMA) immunohistochemistry, and the hydroxyproline (HYP) content revealed similar liver fibrosis levels between KO and WT mice injected intraperitoneally with CCl4. Immunohistochemistry analysis suggested that HIF-2α was mainly expressed in the portal area and hepatic sinusoids but not in hepatocytes. Bioinformatics analyses further indicated that HIF-2α expression was neither liver specific nor hepatocyte specific, and the effect of HIF-2α in hepatocytes on liver fibrosis may not be as important as that in liver sinuses.
Conclusions
Hepatocyte HIF-2α expression may not be a key factor in the initiation of liver fibrogenesis, and hepatocyte-specific deletion of HIF-2α may not be the ideal therapeutic strategy for liver fibrosis.
Collapse
Affiliation(s)
- Jianfang Ye
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
- Molecular Imaging Center, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Jie Chen
- Department of Gastroenterology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yun Li
- Molecular Imaging Center, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Liao Sun
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Hongyun Lu
- Department of Endocrinology and Metabolism, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, Gangdong, China
| |
Collapse
|
22
|
Cannito S, Dianzani U, Parola M, Albano E, Sutti S. Inflammatory processes involved in NASH-related hepatocellular carcinoma. Biosci Rep 2023; 43:BSR20221271. [PMID: 36691794 PMCID: PMC9874450 DOI: 10.1042/bsr20221271] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/09/2022] [Accepted: 01/05/2023] [Indexed: 01/25/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related death worldwide. In the recent years nonalcoholic fatty liver disease (NAFLD) is becoming a growing cause of HCCs and the incidence of NAFLD-related HCCs is expected to further dramatically increase by the next decade. Chronic inflammation is regarded as the driving force of NAFLD progression and a key factor in hepatic carcinogenesis. Hepatic inflammation in NAFLD results from the persistent stimulation of innate immunity in response to hepatocellular injury and gut dysbiosis as well as by the activation of adaptive immunity. However, the relative roles of innate and adaptive immunity in the processes leading to HCC are still incompletely characterized. This is due to the complex interplay between different liver cell populations, which is also strongly influenced by gut-derived bacterial products, metabolic/nutritional signals. Furthermore, carcinogenic mechanisms in NAFLD/NASH appear to involve the activation of signals mediated by hypoxia inducible factors. This review discusses recent data regarding the contribution of different inflammatory cells to NAFLD-related HCC and their possible impact on patient response to current treatments.
Collapse
Affiliation(s)
- Stefania Cannito
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Turin, Turin, Italy
| | - Umberto Dianzani
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| | - Maurizio Parola
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Turin, Turin, Italy
| | - Emanuele Albano
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| | - Salvatore Sutti
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| |
Collapse
|
23
|
Remignon H, Burgues P. Evolution of oxidative stress markers in livers of ducks during force-feeding. Sci Rep 2023; 13:1046. [PMID: 36658173 PMCID: PMC9852233 DOI: 10.1038/s41598-022-27271-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/29/2022] [Indexed: 01/20/2023] Open
Abstract
Mule ducks have been force-fed to develop a hepatic steatosis, also called "foie gras", which is similar to the non-alcoholic fatty liver disease (NAFLD) described in humans and mammals. However, in hepatic steatosis resulting from force-feeding of ducks, very little is known about the fine biochemical events that occur due to the enormous and very rapid increase in total lipids that mainly accumulate in hepatocytes. To begin to reduce this lack of knowledge associated with the development of this specific hepatic steatosis, liver samples were taken at different times to follow the overall biochemical transformation of the liver as well as different markers of oxidative stress, hypoxia and apoptosis. The results indicate that the lipid content increases rapidly in the liver throughout the force-feeding period while the protein content decreases. The amount of hydroxyproline remains constant indicating that no liver fibrosis develops during the force-feeding period. On the contrary, all the tested biomarkers of cellular oxidative stress increase rapidly but without any visible disorder in the coordination of paired activities. At the same time, hypoxia-inducible factors also increase indicating that a hypoxia situation is gradually occurring in hepatocytes. This leads, in addition to the lipotoxicity induced by the accumulation of lipids, to an increased number of liver cells to enter into apoptosis. A relative variability in the level of these cellular responses was also observed indicating that, probably, certain animals support the development of this steatosis differently. This leads us to imagine that the physiological status of these birds may differ widely for reasons that remain to be clarified.
Collapse
Affiliation(s)
- Herve Remignon
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, UPS, 31300, Toulouse, France. .,INP-ENSAT, Université de Toulouse, 31320, Castanet-Tolosan, France.
| | - Pierre Burgues
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, UPS, 31300, Toulouse, France
| |
Collapse
|
24
|
Wu MF, Zhang GD, Liu TT, Shen JH, Cheng JL, Shen J, Yang TY, Huang C, Zhang L. Hif-2α regulates lipid metabolism in alcoholic fatty liver disease through mitophagy. Cell Biosci 2022; 12:198. [PMID: 36476627 PMCID: PMC9730692 DOI: 10.1186/s13578-022-00889-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 08/24/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Disordered lipid metabolism plays an essential role in both the initiation and progression of alcoholic fatty liver disease (AFLD), and fatty acid β-oxidation is increasingly considered as a crucial factor for controlling lipid metabolism. Hif-2α is a member of the Hif family of nuclear receptors, which take part in regulating hepatic fatty acid β-oxidation. However, its functional role in AFLD and the underlying mechanisms remain unclear. RESULTS Hif-2α was upregulated in EtOH-fed mice and EtOH-treated AML-12 cells. Inhibition or silencing of Hif-2α led to increased fatty acid β-oxidation and BNIP3-dependent mitophagy. Downregulation of Hif-2α activates the PPAR-α/PGC-1α signaling pathway, which is involved in hepatic fatty acid β-oxidation, by mediating BNIP3-dependent mitophagy, ultimately delaying the progression of AFLD. CONCLUSIONS Hif-2α induces liver steatosis, which promotes the progression of AFLD. Here, we have described a novel Hif-2α-BNIP3-dependent mitophagy regulatory pathway interconnected with EtOH-induced lipid accumulation, which could be a potential therapeutic target for the prevention and treatment of AFLD.
Collapse
Affiliation(s)
- Mei-fei Wu
- grid.186775.a0000 0000 9490 772XSchool of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032 China ,grid.16821.3c0000 0004 0368 8293Sixth People’s Hospital South Campus, Shanghai Jiaotong University, Shanghai, 201400 China
| | - Guo-dong Zhang
- grid.186775.a0000 0000 9490 772XSchool of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032 China ,grid.186775.a0000 0000 9490 772XThe Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032 China
| | - Tong-tong Liu
- grid.186775.a0000 0000 9490 772XSchool of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032 China ,grid.186775.a0000 0000 9490 772XThe Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032 China
| | - Jun-hao Shen
- grid.186775.a0000 0000 9490 772XSchool of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032 China ,grid.186775.a0000 0000 9490 772XThe Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032 China
| | - Jie-ling Cheng
- grid.186775.a0000 0000 9490 772XSchool of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032 China ,grid.186775.a0000 0000 9490 772XThe Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032 China
| | - Jie Shen
- grid.186775.a0000 0000 9490 772XSchool of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032 China ,grid.186775.a0000 0000 9490 772XThe Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032 China
| | - Tian-yu Yang
- grid.186775.a0000 0000 9490 772XSchool of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032 China ,grid.186775.a0000 0000 9490 772XThe Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032 China
| | - Cheng Huang
- grid.186775.a0000 0000 9490 772XSchool of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032 China ,grid.186775.a0000 0000 9490 772XThe Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032 China
| | - Lei Zhang
- grid.186775.a0000 0000 9490 772XSchool of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032 China ,grid.186775.a0000 0000 9490 772XThe Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032 China
| |
Collapse
|
25
|
Ren X, Diao X, Zhuang J, Wu D. Structural basis for the allosteric inhibition of hypoxia-inducible factor (HIF)-2 by belzutifan. Mol Pharmacol 2022; 102:MOLPHARM-AR-2022-000525. [PMID: 36167425 DOI: 10.1124/molpharm.122.000525] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 08/19/2022] [Accepted: 09/07/2022] [Indexed: 11/22/2022] Open
Abstract
Hypoxia-inducible factor (HIF)-2α and its obligate heterodimerization partner aryl hydrocarbon receptor nuclear translocator (ARNT), are both members of the basic helix-loop-helix-PER-ARNT-SIM (bHLH-PAS) transcription factor family. Previous studies have identified HIF-2α as a key oncogenic driver in clear cell renal cell carcinoma (ccRCC), rendering it a promising drug target for this type of kidney cancer. Belzutifan is the first HIF-2α inhibitor approved for treating ccRCC and other cancers associated with the von Hippel-Lindau (VHL) disease. However, the detailed inhibitory mechanism of belzutifan at molecular level is still unclear. Here we obtained the crystal structure of HIF-2α-ARNT heterodimer in complex with belzutifan at 2.75 Å resolution. The complex structure shows that belzutifan binds into the PAS-B pocket of HIF-2α, and it destabilizes the dimerization of HIF-2α and ARNT through allosteric effects mainly mediated by the key residue M252 of HIF-2α near the dimer interface. We further explored the inhibitory effects of belzutifan using biochemical and functional assays. The time-resolved fluorescence energy transfer (TR-FRET)-based binding assay showed that belzutifan disrupts the dimerization of HIF-2α and ARNT with a Ki value of 20 nM. The luciferase reporter assay indicated that belzutifan can efficiently inhibit the transcriptional activity of HIF-2α with an IC50 value of 17 nM. Besides, the real-time PCR assay illustrated that belzutifan can reduce the expression of HIF-2α downstream genes in 786-O kidney cancer cells in a dose-dependent manner. Our work reveals the molecular mechanism by which belzutifan allosterically inhibits HIF-2α and provides valuable information for the subsequent drug development targeting HIF-2α. Significance Statement The bHLH-PAS family of transcription factors are an emerging group of small-molecule drug targets. Belzutifan, originally developed by Peloton Therapeutics, is the first FDA-approved drug directly binding to a bHLH-PAS protein, the hypoxia-inducible factor (HIF)-2α. Based on the protein-drug complex structure, biochemical binding assays, and functional profiling of downstream gene expression, this study reveals the regulatory mechanism of how belzutifan allosterically destabilizes HIF-2α's heterodimerization with its obligate partner protein, thus reducing their transcriptional activity that links to tumor progression.
Collapse
Affiliation(s)
- Xintong Ren
- State Key Laboratory of Microbial Technology, Shandong University, China
| | - Xiaotong Diao
- State Key Laboratory of Microbial Technology, Shandong University, China
| | | | - Dalei Wu
- State Key Laboratory of Microbial Technology, Shandong University, China
| |
Collapse
|
26
|
Gao H, Jin Z, Bandyopadhyay G, Wang G, Zhang D, Rocha KCE, Liu X, Zhao H, Kisseleva T, Brenner DA, Karin M, Ying W. Aberrant iron distribution via hepatocyte-stellate cell axis drives liver lipogenesis and fibrosis. Cell Metab 2022; 34:1201-1213.e5. [PMID: 35921818 PMCID: PMC9365100 DOI: 10.1016/j.cmet.2022.07.006] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/11/2022] [Accepted: 07/13/2022] [Indexed: 12/11/2022]
Abstract
Hepatocytes have important roles in liver iron homeostasis, abnormalities in which are tightly associated with liver steatosis and fibrosis. Here, we show that non-alcoholic fatty liver disease (NAFLD) and steatohepatitis (NASH) are characterized by iron-deficient hepatocytes and iron overload in hepatic stellate cells (HSCs). Iron deficiency enhances hepatocyte lipogenesis and insulin resistance through HIF2α-ATF4 signaling. Elevated secretion of iron-containing hepatocyte extracellular vesicles (EVs), which are normally cleared by Kupffer cells, accounts for hepatocyte iron deficiency and HSC iron overload in NAFLD/NASH livers. Iron accumulation results in overproduction of reactive oxygen species that promote HSC fibrogenic activation. Conversely, blocking hepatocyte EV secretion or depleting EV iron cargo restores liver iron homeostasis, concomitant with mitigation of NAFLD/NASH-associated liver steatosis and fibrosis. Taken together, these studies show that iron distribution disorders contribute to the development of liver metabolic diseases.
Collapse
Affiliation(s)
- Hong Gao
- Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| | - Zhongmou Jin
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Gautam Bandyopadhyay
- Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Gaowei Wang
- Department of Pediatrics, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA, USA
| | - Dinghong Zhang
- Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Karina Cunha E Rocha
- Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Xiao Liu
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Surgery, University of California, San Diego, La Jolla, CA, USA
| | - Huayi Zhao
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Tatiana Kisseleva
- Department of Surgery, University of California, San Diego, La Jolla, CA, USA
| | - David A Brenner
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Michael Karin
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA.
| | - Wei Ying
- Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
27
|
Novo E, Cappon A, Villano G, Quarta S, Cannito S, Bocca C, Turato C, Guido M, Maggiora M, Protopapa F, Sutti S, Provera A, Ruvoletto M, Biasiolo A, Foglia B, Albano E, Pontisso P, Parola M. SerpinB3 as a Pro-Inflammatory Mediator in the Progression of Experimental Non-Alcoholic Fatty Liver Disease. Front Immunol 2022; 13:910526. [PMID: 35874657 PMCID: PMC9304805 DOI: 10.3389/fimmu.2022.910526] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is becoming the most common chronic liver disease worldwide. In 20-30% of patients, NAFLD can progress into non-alcoholic steatohepatitis (NASH), eventually leading to fibrosis, cirrhosis and hepatocellular carcinoma development. SerpinB3 (SB3), a hypoxia-inducible factor-2α dependent cysteine protease inhibitor, is up-regulated in hepatocytes during progressive NAFLD and proposed to contribute to disease progression. In this study we investigated the pro-inflammatory role of SB3 by employing phorbol-myristate acetate-differentiated human THP-1 macrophages exposed in vitro to human recombinant SB3 (hrSB3) along with mice overexpressing SB3 in hepatocytes (TG/SB3) or knockout for SB3 (KO/SB3) in which NASH was induced by feeding methionine/choline deficient (MCD) or a choline-deficient, L-amino acid defined (CDAA) diets. In vivo experiments showed that the induction of NASH in TG/SB3 mice was characterized by an impressive increase of liver infiltrating macrophages that formed crown-like aggregates and by an up-regulation of hepatic transcript levels of pro-inflammatory cytokines. All these parameters and the extent of liver damage were significantly blunted in KO/SB3 mice. In vitro experiments confirmed that hrSB3 stimulated macrophage production of M1-cytokines such as TNFα and IL-1β and reactive oxygen species along with that of TGFβ and VEGF through the activation of the NF-kB transcription factor. The opposite changes in liver macrophage activation observed in TG/SB3 or KO/SB3 mice with NASH were associated with a parallel modulation in the expression of triggering receptor expressed on myeloid cells-2 (TREM2), CD9 and galectin-3 markers, recently detected in NASH-associated macrophages. From these results we propose that SB3, produced by activated/injured hepatocytes, may operate as a pro-inflammatory mediator in NASH contributing to the disease progression.
Collapse
Affiliation(s)
- Erica Novo
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Andrea Cappon
- Department of Medicine, University of Padova, Padova, Italy
| | - Gianmarco Villano
- Department of Surgical, Oncological and Gastroenterological Sciences – DISCOG, University of Padova, Padova, Italy
| | - Santina Quarta
- Department of Medicine, University of Padova, Padova, Italy
| | - Stefania Cannito
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Claudia Bocca
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Cristian Turato
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Maria Guido
- Department of Medicine, University of Padova, Padova, Italy
| | - Marina Maggiora
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Francesca Protopapa
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Salvatore Sutti
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Alessia Provera
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | | | | | - Beatrice Foglia
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Emanuele Albano
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | | | - Maurizio Parola
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| |
Collapse
|
28
|
Hypoxia signaling in human health and diseases: implications and prospects for therapeutics. Signal Transduct Target Ther 2022; 7:218. [PMID: 35798726 PMCID: PMC9261907 DOI: 10.1038/s41392-022-01080-1] [Citation(s) in RCA: 198] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 02/07/2023] Open
Abstract
Molecular oxygen (O2) is essential for most biological reactions in mammalian cells. When the intracellular oxygen content decreases, it is called hypoxia. The process of hypoxia is linked to several biological processes, including pathogenic microbe infection, metabolic adaptation, cancer, acute and chronic diseases, and other stress responses. The mechanism underlying cells respond to oxygen changes to mediate subsequent signal response is the central question during hypoxia. Hypoxia-inducible factors (HIFs) sense hypoxia to regulate the expressions of a series of downstream genes expression, which participate in multiple processes including cell metabolism, cell growth/death, cell proliferation, glycolysis, immune response, microbe infection, tumorigenesis, and metastasis. Importantly, hypoxia signaling also interacts with other cellular pathways, such as phosphoinositide 3-kinase (PI3K)-mammalian target of rapamycin (mTOR) signaling, nuclear factor kappa-B (NF-κB) pathway, extracellular signal-regulated kinases (ERK) signaling, and endoplasmic reticulum (ER) stress. This paper systematically reviews the mechanisms of hypoxia signaling activation, the control of HIF signaling, and the function of HIF signaling in human health and diseases. In addition, the therapeutic targets involved in HIF signaling to balance health and diseases are summarized and highlighted, which would provide novel strategies for the design and development of therapeutic drugs.
Collapse
|
29
|
Chronic intermittent hypoxia contributes to non-alcoholic steatohepatitis progression in patients with obesity. Hepatol Int 2022; 16:824-834. [DOI: 10.1007/s12072-022-10347-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/22/2022] [Indexed: 11/04/2022]
|
30
|
Hypoxia-driven metabolic heterogeneity and immune evasive behaviour of gastrointestinal cancers: Elements of a recipe for disaster. Cytokine 2022; 156:155917. [PMID: 35660715 DOI: 10.1016/j.cyto.2022.155917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/28/2022] [Accepted: 05/16/2022] [Indexed: 11/24/2022]
Abstract
Gastrointestinal (GI) cancers refer to a group of malignancies associated with the GI tract (GIT). Like other solid tumors, hypoxic regions consistently feature inside the GI tumor microenvironment (TME) and contribute towards metabolic reprogramming of tumor-resident cells by modulating hypoxia-induced factors. We highlight here how the metabolic crosstalk between cancer cells and immune cells generate immunosuppressive environment inside hypoxic tumors. Given the fluctuating nature of tumor hypoxia, the metabolic fluxes between immune cells and cancer cells change dynamically. These changes alter cellular phenotypes and functions, resulting in the acceleration of cancer progression. These evolved properties of hypoxic tumors make metabolism-targeting monotherapy approaches or immunotherapy-measures unsuccessful. The current review highlights the advantages of combined immunometabolic treatment strategies to target hypoxic GI cancers and also identifies research areas to develop better combinational therapeutics for future.
Collapse
|
31
|
Di Maira G, Foglia B, Napione L, Turato C, Maggiora M, Sutti S, Novo E, Alvaro M, Autelli R, Colombatto S, Bussolino F, Carucci P, Gaia S, Rosso C, Biasiolo A, Pontisso P, Bugianesi E, Albano E, Marra F, Parola M, Cannito S. Oncostatin M is overexpressed in NASH-related hepatocellular carcinoma and promotes cancer cell invasiveness and angiogenesis. J Pathol 2022; 257:82-95. [PMID: 35064579 PMCID: PMC9315146 DOI: 10.1002/path.5871] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/03/2021] [Accepted: 01/13/2022] [Indexed: 12/03/2022]
Abstract
Oncostatin M (OSM) is a pleiotropic cytokine of the interleukin (IL)-6 family that contributes to the progression of chronic liver disease. Here we investigated the role of OSM in the development and progression of hepatocellular carcinoma (HCC) in non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH). The role of OSM was investigated in (1) selected cohorts of NAFLD/NASH HCC patients, (2) liver cancer cells exposed to human recombinant OSM or stably transfected to overexpress human OSM, (3) murine HCC xenografts, and (4) a murine NASH-related model of hepatic carcinogenesis. OSM was found to be selectively overexpressed in HCC cells of NAFLD/NASH patients, depending on tumor grade. OSM serum levels, barely detectable in patients with simple steatosis or NASH, were increased in patients with cirrhosis and more evident in those carrying HCC. In this latter group, OSM serum levels were significantly higher in the subjects with intermediate/advanced HCCs and correlated with poor survival. Cell culture experiments indicated that OSM upregulation in hepatic cancer cells contributes to HCC progression by inducing epithelial-to-mesenchymal transition and increased invasiveness of cancer cells as well as by inducing angiogenesis, which is of critical relevance. In murine xenografts, OSM overexpression was associated with slower tumor growth but an increased rate of lung metastases. Overexpression of OSM and its positive correlation with the angiogenic switch were also confirmed in a murine model of NAFLD/NASH-related hepatocarcinogenesis. Consistent with this, analysis of liver specimens from human NASH-related HCCs with vascular invasion showed that OSM was expressed by liver cancer cells invading hepatic vessels. In conclusion, OSM upregulation appears to be a specific feature of HCC arising on a NAFLD/NASH background, and it correlates with clinical parameters and disease outcome. Our data highlight a novel pro-carcinogenic contribution for OSM in NAFLD/NASH, suggesting a role of this factor as a prognostic marker and a putative potential target for therapy. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Giovanni Di Maira
- Department of Clinical and Experimental Medicine and Center DenotheUniversity of FirenzeFirenzeItaly
| | - Beatrice Foglia
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical PathologyUniversity of TorinoTorinoItaly
| | - Lucia Napione
- Laboratory of Vascular Oncology Candiolo Cancer Institute – FPO IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico)TorinoItaly
- Department of Applied Science and TechnologyPolitecnico di TorinoTorinoItaly
| | - Cristian Turato
- Department of Molecular MedicineUniversity of PaviaPaviaItaly
| | - Marina Maggiora
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical PathologyUniversity of TorinoTorinoItaly
| | - Salvatore Sutti
- Department of Health Sciences and Interdisciplinary Research Center for Autoimmune DiseasesUniversity Amedeo Avogadro of East PiedmontNovaraItaly
| | - Erica Novo
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical PathologyUniversity of TorinoTorinoItaly
| | - Maria Alvaro
- Laboratory of Vascular Oncology Candiolo Cancer Institute – FPO IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico)TorinoItaly
- Department of OncologyUniversity of TorinoTorinoItaly
| | - Riccardo Autelli
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical PathologyUniversity of TorinoTorinoItaly
| | | | - Federico Bussolino
- Laboratory of Vascular Oncology Candiolo Cancer Institute – FPO IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico)TorinoItaly
- Department of OncologyUniversity of TorinoTorinoItaly
| | - Patrizia Carucci
- Division of GastroenterologyCittà della Salute e della Scienza University‐HospitalTurinItaly
| | - Silvia Gaia
- Division of GastroenterologyCittà della Salute e della Scienza University‐HospitalTurinItaly
| | - Chiara Rosso
- Department of Medical SciencesUniversity of TorinoTorinoItaly
| | | | | | | | - Emanuele Albano
- Department of Health Sciences and Interdisciplinary Research Center for Autoimmune DiseasesUniversity Amedeo Avogadro of East PiedmontNovaraItaly
| | - Fabio Marra
- Department of Clinical and Experimental Medicine and Center DenotheUniversity of FirenzeFirenzeItaly
| | - Maurizio Parola
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical PathologyUniversity of TorinoTorinoItaly
| | - Stefania Cannito
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical PathologyUniversity of TorinoTorinoItaly
| |
Collapse
|
32
|
Landete P, Fernández-García CE, Aldave-Orzaiz B, Hernández-Olivo M, Acosta-Gutiérrez CM, Zamora-García E, Ancochea J, González-Rodríguez Á, García-Monzón C. Increased Oxygen Desaturation Time During Sleep Is a Risk Factor for NASH in Patients With Obstructive Sleep Apnea: A Prospective Cohort Study. Front Med (Lausanne) 2022; 9:808417. [PMID: 35280896 PMCID: PMC8906568 DOI: 10.3389/fmed.2022.808417] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Given that obstructive sleep apnea (OSA) is commonly associated with metabolic disorders, in this prospective study, we sought to determine the prevalence and risk factors for hepatosteatosis, non-alcoholic steatohepatitis (NASH), and advanced liver fibrosis in patients with clinical and polygraphic criteria of OSA (n = 153) and in subjects with normal lung function parameters (NLP, n = 43). Methods Hepatosteatosis, NASH, and advanced liver fibrosis were determined by blood-based non-invasive tools, such as the fatty liver index and the hepatic steatosis index, a serum lipidomic (OWLiver™) test, and three distinct fibrosis algorithms, respectively. Logistic regression models adjusted by potential confounders were performed to evaluate risk factors. Results Insulin resistance and dyslipidemia were more frequent in patients with OSA than in subjects with NLP. The prevalence of hepatosteatosis was significantly higher in patients with OSA than in subjects with NLP. NASH was also found more frequently in patients with OSA than in subjects with NLP. In contrast, advanced liver fibrosis was rarely detected in the entire study population, and no significant differences were observed between patients with OSA and subjects with NLP. Besides male gender, increased body mass index (BMI), and presence of type 2 diabetes, percentage of sleep time with oxygen saturation <90% (Tc90%) was the only polygraphic variable significantly associated with NASH in patients with OSA. Conclusions This study shows that hepatosteatosis and NASH are highly prevalent in patients with OSA and indicates that those with a Tc90% higher than 10% are at increased risk for NASH.
Collapse
Affiliation(s)
- Pedro Landete
- Servicio de Neumología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain.,Departamento de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos Ernesto Fernández-García
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain
| | - Beatriz Aldave-Orzaiz
- Servicio de Neumología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain
| | - Marta Hernández-Olivo
- Servicio de Neumología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain
| | - Carmen M Acosta-Gutiérrez
- Servicio de Neumología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain
| | - Enrique Zamora-García
- Servicio de Neumología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain.,Departamento de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Julio Ancochea
- Servicio de Neumología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain.,Departamento de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Águeda González-Rodríguez
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Carmelo García-Monzón
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain
| |
Collapse
|
33
|
Pan Y, Deng L, Wang H, He K, Xia Q. Histidine-rich glycoprotein (HRGP): Pleiotropic and paradoxical effects on macrophage, tumor microenvironment, angiogenesis, and other physiological and pathological processes. Genes Dis 2022; 9:381-392. [PMID: 35224154 PMCID: PMC8843877 DOI: 10.1016/j.gendis.2020.07.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/15/2020] [Accepted: 07/31/2020] [Indexed: 12/14/2022] Open
Abstract
Histidine-rich glycoprotein (HRGP) is a relatively less known glycoprotein, but it is abundant in plasma with a multidomain structure, which allows it to interact with many ligands and regulate various biological processes. HRGP ligands includes heme, Zn2+, thrombospondin, plasmin/plasminogen, heparin/heparan sulfate, fibrinogen, tropomyosin, IgG, FcγR, C1q. In many conditions, the histidine-rich region of HRGP strengthens ligand binding following interaction with Zn2+ or exposure to low pH, such as sites of tissue injury or tumor growth. The multidomain structure and diverse ligand binding attributes of HRGP indicates that it can act as an extracellular adaptor protein, connecting with different ligands, especially on cell surfaces. Also, HRGP can selectively target IgG, which blocks the production of soluble immune complexes. The most common cell surface ligand of HRGP is heparan sulfate proteoglycan, and the interaction is also potentiated by elevated Zn2+ concentration and low pH. Recent reports have shown that HRGP can modulate macrophage polarization and possibly regulate other physiological processes such as angiogenesis, anti-tumor immune response, fibrinolysis and coagulation, soluble immune complex clearance and phagocytosis of apoptotic/necrosis cells. In addition, it has also been reported that HRGP has antibacterial and anti-HIV infection effects and may be used as a novel clinical biomarker accordingly. This review outlines the molecular, structural and biological properties of HRGP as well as presenting an update on the function of HRGP in various physiological processes.
Collapse
|
34
|
Cannito S, Bincoletto V, Turato C, Pontisso P, Scupoli MT, Ailuno G, Andreana I, Stella B, Arpicco S, Bocca C. Hyaluronated and PEGylated Liposomes as a Potential Drug-Delivery Strategy to Specifically Target Liver Cancer and Inflammatory Cells. Molecules 2022; 27:molecules27031062. [PMID: 35164326 PMCID: PMC8840578 DOI: 10.3390/molecules27031062] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 11/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most frequent primary liver cancer and is characterized by poor clinical outcomes, with the majority of patients not being eligible for curative therapy and treatments only being applicable for early-stage tumors. CD44 is a receptor for hyaluronic acid (HA) and is involved in HCC progression. The aim of this work is to propose HA- and PEGylated-liposomes as promising approaches for the treatment of HCC. It has been found, in this work, that CD44 transcripts are up-regulated in HCC patients, as well as in a murine model of NAFLD/NASH-related hepatocarcinogenesis. Cell culture experiments indicate that HA-liposomes are more rapidly and significantly internalized by Huh7 cells that over-express CD44, compared with HepG2 cells that express low levels of the receptor, in which the uptake seems due to endocytic events. By contrast, human and murine macrophage cell lines (THP-1, RAW264.7) show improved and rapid uptake of PEG-modified liposomes without the involvement of the CD44. Moreover, the internalization of PEG-modified liposomes seems to induce polarization of THP1 towards the M1 phenotype. In conclusion, data reported in this study indicate that this strategy can be proposed as an alternative for drug delivery and one that dually and specifically targets liver cancer cells and infiltrating tumor macrophages in order to counteract two crucial aspect of HCC progression.
Collapse
Affiliation(s)
- Stefania Cannito
- Department of Clinical and Biological Sciences, University of Turin, 10125 Turin, Italy; (S.C.); (C.B.)
| | - Valeria Bincoletto
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (V.B.); (I.A.); (B.S.)
| | - Cristian Turato
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
| | | | - Maria Teresa Scupoli
- Research Center LURM, Interdepartmental Laboratory of Medical Research, University of Verona, 37134 Verona, Italy;
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37129 Verona, Italy
| | - Giorgia Ailuno
- Department of Pharmacy, University of Genova, 16148 Genova, Italy;
| | - Ilaria Andreana
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (V.B.); (I.A.); (B.S.)
| | - Barbara Stella
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (V.B.); (I.A.); (B.S.)
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (V.B.); (I.A.); (B.S.)
- Correspondence: ; Tel.: +39-011-670-6668
| | - Claudia Bocca
- Department of Clinical and Biological Sciences, University of Turin, 10125 Turin, Italy; (S.C.); (C.B.)
| |
Collapse
|
35
|
Li X, Zhang Q, Wang Z, Zhuang Q, Zhao M. Immune and Metabolic Alterations in Liver Fibrosis: A Disruption of Oxygen Homeostasis? Front Mol Biosci 2022; 8:802251. [PMID: 35187072 PMCID: PMC8850363 DOI: 10.3389/fmolb.2021.802251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/17/2021] [Indexed: 12/06/2022] Open
Abstract
According to the WHO, “cirrhosis of the liver” was the 11th leading cause of death globally in 2019. Many kinds of liver diseases can develop into liver cirrhosis, and liver fibrosis is the main pathological presentation of different aetiologies, including toxic damage, viral infection, and metabolic and genetic diseases. It is characterized by excessive synthesis and decreased decomposition of extracellular matrix (ECM). Hepatocyte cell death, hepatic stellate cell (HSC) activation, and inflammation are crucial incidences of liver fibrosis. The process of fibrosis is also closely related to metabolic and immune disorders, which are usually induced by the destruction of oxygen homeostasis, including mitochondrial dysfunction, oxidative stress, and hypoxia pathway activation. Mitochondria are important organelles in energy generation and metabolism. Hypoxia-inducible factors (HIFs) are key factors activated when hypoxia occurs. Both are considered essential factors of liver fibrosis. In this review, the authors highlight the impact of oxygen imbalance on metabolism and immunity in liver fibrosis as well as potential novel targets for antifibrotic therapies.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Quyan Zhang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zeyu Wang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Quan Zhuang
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Quan Zhuang, ; Mingyi Zhao,
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Quan Zhuang, ; Mingyi Zhao,
| |
Collapse
|
36
|
Clavería-Cabello A, Avila MA. HIF2α Activation in NASH: A New Force Pushing Toward HCC. Cell Mol Gastroenterol Hepatol 2021; 13:678-680. [PMID: 34848185 PMCID: PMC8777000 DOI: 10.1016/j.jcmgh.2021.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 10/31/2021] [Accepted: 11/03/2021] [Indexed: 12/13/2022]
Affiliation(s)
| | - Matías A Avila
- Hepatology Program, CIMA, University of Navarra, Pamplona, Spain; CIBERehd, Instituto de Salud Carlos III, Madrid, Spain; Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain.
| |
Collapse
|
37
|
Kotlyarov S, Bulgakov A. Lipid Metabolism Disorders in the Comorbid Course of Nonalcoholic Fatty Liver Disease and Chronic Obstructive Pulmonary Disease. Cells 2021; 10:2978. [PMID: 34831201 PMCID: PMC8616072 DOI: 10.3390/cells10112978] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/25/2021] [Accepted: 10/30/2021] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is currently among the most common liver diseases. Unfavorable data on the epidemiology of metabolic syndrome and obesity have increased the attention of clinicians and researchers to the problem of NAFLD. The research results allow us to emphasize the systemicity and multifactoriality of the pathogenesis of liver parenchyma lesion. At the same time, many aspects of its classification, etiology, and pathogenesis remain controversial. Local and systemic metabolic disorders are also a part of the pathogenesis of chronic obstructive pulmonary disease and can influence its course. The present article analyzes the metabolic pathways mediating the links of impaired lipid metabolism in NAFLD and chronic obstructive pulmonary disease (COPD). Free fatty acids, cholesterol, and ceramides are involved in key metabolic and inflammatory pathways underlying the pathogenesis of both diseases. Moreover, inflammation and lipid metabolism demonstrate close links in the comorbid course of NAFLD and COPD.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia;
| | | |
Collapse
|
38
|
Foglia B, Sutti S, Cannito S, Rosso C, Maggiora M, Autelli R, Novo E, Bocca C, Villano G, Ramavath NN, Younes R, Tusa I, Rovida E, Pontisso P, Bugianesi E, Albano E, Parola M. Hepatocyte-Specific Deletion of HIF2α Prevents NASH-Related Liver Carcinogenesis by Decreasing Cancer Cell Proliferation. Cell Mol Gastroenterol Hepatol 2021; 13:459-482. [PMID: 34655812 PMCID: PMC8688724 DOI: 10.1016/j.jcmgh.2021.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 10/06/2021] [Accepted: 10/06/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND & AIMS Hypoxia and hypoxia-inducible factors (HIFs) are involved in chronic liver disease progression. We previously showed that hepatocyte HIF-2α activation contributed significantly to nonalcoholic fatty liver disease progression in experimental animals and human patients. In this study, using an appropriate genetic murine model, we mechanistically investigated the involvement of hepatocyte HIF-2α in experimental nonalcoholic steatohepatitis (NASH)-related carcinogenesis. METHODS The role of HIF-2α was investigated by morphologic, cellular, and molecular biology approaches in the following: (1) mice carrying hepatocyte-specific deletion of HIF-2α (HIF-2α-/- mice) undergoing a NASH-related protocol of hepatocarcinogenesis; (2) HepG2 cells stably transfected to overexpress HIF-2α; and (3) liver specimens from NASH patients with hepatocellular carcinoma. RESULTS Mice carrying hepatocyte-specific deletion of HIF-2α (hHIF-2α-/-) showed a significant decrease in the volume and number of liver tumors compared with wild-type littermates. These effects did not involve HIF-1α changes and were associated with a decrease of cell proliferation markers proliferating cell nuclear antigen and Ki67. In both human and rodent nonalcoholic fatty liver disease-related tumors, HIF-2α levels were strictly associated with hepatocyte production of SerpinB3, a mediator previously shown to stimulate liver cancer cell proliferation through the Hippo/Yes-associated protein (YAP)/c-Myc pathway. Consistently, we observed positive correlations between the transcripts of HIF-2α, YAP, and c-Myc in individual hepatocellular carcinoma tumor masses, while HIF-2α deletion down-modulated c-Myc and YAP expression without affecting extracellular signal-regulated kinase 1/2, c-Jun N-terminal kinase, and AKT-dependent signaling. In vitro data confirmed that HIF-2α overexpression induced HepG2 cell proliferation through YAP-mediated mechanisms. CONCLUSIONS These results indicate that the activation of HIF-2α in hepatocytes has a critical role in liver carcinogenesis during NASH progression, suggesting that HIF-2α-blocking agents may serve as novel putative therapeutic tools.
Collapse
Affiliation(s)
- Beatrice Foglia
- Unit of Experimental Medicine and Clinical Pathology, Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Salvatore Sutti
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University Amedeo Avogadro of East Piedmont, Novara, Italy
| | - Stefania Cannito
- Unit of Experimental Medicine and Clinical Pathology, Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Chiara Rosso
- Department of Medical Sciences, University of Turin, Torino, Italy; Division of Gastroenterology, San Giovanni Hospital, Torino, Italy
| | - Marina Maggiora
- Unit of Experimental Medicine and Clinical Pathology, Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Riccardo Autelli
- Unit of Experimental Medicine and Clinical Pathology, Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Erica Novo
- Unit of Experimental Medicine and Clinical Pathology, Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Claudia Bocca
- Unit of Experimental Medicine and Clinical Pathology, Department of Clinical and Biological Sciences, University of Turin, Italy
| | | | - Naresh Naik Ramavath
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University Amedeo Avogadro of East Piedmont, Novara, Italy
| | - Ramy Younes
- Department of Medical Sciences, University of Turin, Torino, Italy; Division of Gastroenterology, San Giovanni Hospital, Torino, Italy
| | - Ignazia Tusa
- Unit of Experimental Oncology and Pathology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Elisabetta Rovida
- Unit of Experimental Oncology and Pathology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | | | - Elisabetta Bugianesi
- Department of Medical Sciences, University of Turin, Torino, Italy; Division of Gastroenterology, San Giovanni Hospital, Torino, Italy
| | - Emanuele Albano
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University Amedeo Avogadro of East Piedmont, Novara, Italy
| | - Maurizio Parola
- Unit of Experimental Medicine and Clinical Pathology, Department of Clinical and Biological Sciences, University of Turin, Italy.
| |
Collapse
|
39
|
Holzner LMW, Murray AJ. Hypoxia-Inducible Factors as Key Players in the Pathogenesis of Non-alcoholic Fatty Liver Disease and Non-alcoholic Steatohepatitis. Front Med (Lausanne) 2021; 8:753268. [PMID: 34692739 PMCID: PMC8526542 DOI: 10.3389/fmed.2021.753268] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/10/2021] [Indexed: 12/20/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and its more severe form non-alcoholic steatohepatitis (NASH) are a major public health concern with high and increasing global prevalence, and a significant disease burden owing to its progression to more severe forms of liver disease and the associated risk of cardiovascular disease. Treatment options, however, remain scarce, and a better understanding of the pathological and physiological processes involved could enable the development of new therapeutic strategies. One process implicated in the pathology of NAFLD and NASH is cellular oxygen sensing, coordinated largely by the hypoxia-inducible factor (HIF) family of transcription factors. Activation of HIFs has been demonstrated in patients and mouse models of NAFLD and NASH and studies of activation and inhibition of HIFs using pharmacological and genetic tools point toward important roles for these transcription factors in modulating central aspects of the disease. HIFs appear to act in several cell types in the liver to worsen steatosis, inflammation, and fibrosis, but may nevertheless improve insulin sensitivity. Moreover, in liver and other tissues, HIF activation alters mitochondrial respiratory function and metabolism, having an impact on energetic and redox homeostasis. This article aims to provide an overview of current understanding of the roles of HIFs in NAFLD, highlighting areas where further research is needed.
Collapse
Affiliation(s)
| | - Andrew J. Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
40
|
Kinsella GK, Cannito S, Bordano V, Stephens JC, Rosa AC, Miglio G, Guaschino V, Iannaccone V, Findlay JBC, Benetti E. GPR21 Inhibition Increases Glucose-Uptake in HepG2 Cells. Int J Mol Sci 2021; 22:ijms221910784. [PMID: 34639123 PMCID: PMC8509304 DOI: 10.3390/ijms221910784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 02/08/2023] Open
Abstract
GPR21 is a constitutively active, orphan, G-protein-coupled receptor, with in vivo studies suggesting its involvement in the modulation of insulin sensitivity. However, its precise contribution is not fully understood. As the liver is both a major target of insulin signalling and critically involved in glucose metabolism, the aim of this study was to examine the role of GPR21 in the regulation of glucose uptake and production in human hepatocytes. In particular, HepG2 cells, which express GPR21, were adopted as cellular models. Compared with untreated cells, a significant increase in glucose uptake was measured in cells treated with siRNA to downregulate GPR21 expression or with the GPR21-inverse agonist, GRA2. Consistently, a significantly higher membrane translocation of GLUT-2 was measured under these conditions. These effects were accompanied by an increased ratio of phAKT(Ser473)/tot-AKT and phGSK-3β(Ser9)/tot-GSK-3β, thus indicating a marked activation of the insulin signalling pathway. Moreover, a significant reduction in ERK activation was observed with GPR21 inhibition. Collectively, these results indicate that GPR21 mediates the negative effects on glucose uptake by the liver cells. In addition, they suggest that the pharmacological inhibition of GPR21 could be a novel strategy to improve glucose homeostasis and counteract hepatic insulin resistance.
Collapse
Affiliation(s)
- Gemma K. Kinsella
- School of Food Sciences and Environmental Health, Technological University Dublin, Grangegorman, D07 ADY7 Dublin, Ireland;
| | - Stefania Cannito
- Department of Clinical and Biological Sciences, University of Turin, 10125 Turin, Italy;
| | - Valentina Bordano
- Dipartimento di Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.C.R.); (G.M.); (V.G.); (V.I.)
| | - John C. Stephens
- Department of Chemistry, Maynooth University, Maynooth, Co. Kildare, Ireland;
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Arianna C. Rosa
- Dipartimento di Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.C.R.); (G.M.); (V.G.); (V.I.)
| | - Gianluca Miglio
- Dipartimento di Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.C.R.); (G.M.); (V.G.); (V.I.)
| | - Valeria Guaschino
- Dipartimento di Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.C.R.); (G.M.); (V.G.); (V.I.)
| | - Valeria Iannaccone
- Dipartimento di Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.C.R.); (G.M.); (V.G.); (V.I.)
| | - John B. C. Findlay
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland;
- School of Biomedical Sciences, University of Leeds, LS2 9JT Leeds, UK
| | - Elisa Benetti
- Dipartimento di Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.C.R.); (G.M.); (V.G.); (V.I.)
- Correspondence: ; Tel.: +39-0116707137
| |
Collapse
|
41
|
Zhao M, Wang S, Zuo A, Zhang J, Wen W, Jiang W, Chen H, Liang D, Sun J, Wang M. HIF-1α/JMJD1A signaling regulates inflammation and oxidative stress following hyperglycemia and hypoxia-induced vascular cell injury. Cell Mol Biol Lett 2021; 26:40. [PMID: 34479471 PMCID: PMC8414688 DOI: 10.1186/s11658-021-00283-8] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/19/2021] [Indexed: 11/10/2022] Open
Abstract
Background Endothelial cell (EC) injury accelerates the progression of diabetic macrovascular complications. Hypoxia is an important cause of EC injury. Hypoxia-inducible factor-1 alpha (HIF-1α) is an important hypoxia regulatory protein. Our previous studies showed that high-glucose and hypoxic conditions could upregulate HIF-1α expression and enhance EC inflammatory injury, independently of the nuclear factor kappa-B (NF-κB) pathway. However, it is not clear whether HIF-1α plays a role in vascular disease through epigenetic-related mechanisms. Methods We conducted gene expression analysis and molecular mechanistic studies in human umbilical vein endothelial cells (HUVECs) induced by hyperglycemia and hypoxia using RNA sequencing (RNA-seq) and small interfering HIF-1α (si-HIF-1α). We determined HIF-1α and Jumonji domain-containing protein 1 A (JMJD1A) expression by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blot, analyzed inflammatory protein secretion in the cell supernatant by enzymelinked immunosorbent assay (ELISA), and assessed protein interaction between HIF-1α and JMJD1A by chromatin immunoprecipitation (Ch-IP). We used the Cell Counting Kit8 (CCK-8) assay to analyze cell viability, and assessed oxidative stress indicators by using a detection kit and flow cytometry. Results High glucose and hypoxia up-regulated HIF-1α expression, and down-regulated HIF-1α decreased the level of inflammation and oxidative stress in HUVECs. To determine the downstream pathways, we observed histone demethylases genes and related pathway by RNA-sEq. Among these, JMJD1A was the most upregulated gene in histone demethylases. Moreover, we observed that HIF-1α bound to the promoter of JMJD1A, and the ameliorative effects of si-HIF-1α on oxidative stress and inflammatory cytokines in high-glucose and hypoxia-induced HUVECs were reversed by JMJD1A overexpression. Furthermore, knockdown of JMJD1A decreased inflammatory and oxidative stress injury. To determine the JMJD1A-related factors, we conducted gene expression analysis on JMJD1A-knockdown HUVECs. We observed that downregulation of inflammation and the oxidative stress pathway were enriched and FOS and FOSB might be important protective transcription factors. Conclusions These findings provide novel evidence that the HIF-1α/JMJD1A signaling pathway is involved in inflammation and oxidative stress in HUVECs induced by high glucose and hypoxia. Also, this pathway might act as a novel regulator of oxidative stress and inflammatory-related events in response to diabetic vascular injury and thus contribute to the pathological progression of diabetes and vascular disease. Supplementary Information The online version contains supplementary material available at 10.1186/s11658-021-00283-8.
Collapse
Affiliation(s)
- Min Zhao
- Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Shaoting Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Anna Zuo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Jiaxing Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Weiheng Wen
- Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Weiqiang Jiang
- Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Hong Chen
- Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Donghui Liang
- Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Jia Sun
- Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| | - Ming Wang
- Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China. .,School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| |
Collapse
|
42
|
Wang H, Wang L, Li Y, Luo S, Ye J, Lu Z, Li X, Lu H. The HIF-2α/PPARα pathway is essential for liraglutide-alleviated, lipid-induced hepatic steatosis. Biomed Pharmacother 2021; 140:111778. [PMID: 34062416 DOI: 10.1016/j.biopha.2021.111778] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
Liraglutide has been demonstrated to alleviate hepatic steatosis in clinical practice, but the underlying mechanism remains unclear. Our previous study indicated that the HIF-2α/PPARα pathway was involved in hepatic lipid accumulation induced by hypoxia.We aimed to investigate whether liraglutide could alleviate lipid-induced hepatic steatosis via the HIF-2α/PPARα pathway. Whole-body HIF-2α heterozygous knockout (HIF-2α+/-) mice and littermate wild-type (WT) mice were successfully established. Male mice challenged with a high-fat diet were treated with liraglutide (0.6 mg/kg/d) or normal saline by intraperitoneal injection for 4 weeks. We observed that, compared with WT mice, many indicators of HIF-2α+/- mice improved, including GTT, ITT, fasting blood glucose, body weight, liver weight, and lipid profile in serum or liver lipid deposition, and the expression level of PPARα, mitochondrial function genes, and fatty acid oxidation genes were upregulated, while those of HIF-2α and lipogenesis genes were downregulated significantly. After liraglutide treatment in WT mice, we found that significant improvements were observed in the fat mass, GTT, ITT, fasting blood glucose, body weight, liver weight, lipid profile in serum or liver lipid deposition; the β-oxidation genes were upregulated and the lipogenesis genes were downregulated; and the abundance of intestinal Akkermansia muciniphila increased significantly. However, the effects of liraglutide on WT mice were not observed in HIF-2α+/- mice. In addition, in the HepG2 steatotic hepatocyte model, liraglutide alleviated lipid deposits by repressing lipid synthesis and enhancing fatty acid β-oxidation, which were substantially suppressed by the HIF-2α modulators. Therefore, the HIF-2α/PPARα pathway is essential for liraglutide-alleviated lipid-induced hepatic steatosis.
Collapse
Affiliation(s)
- Hou Wang
- Department of Endocrinology & Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China; Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Lingling Wang
- Department of Endocrinology & Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Yun Li
- Department of Endocrinology & Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China; Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Shunkui Luo
- Department of Endocrinology & Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Jianfang Ye
- Department of Endocrinology & Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Zhanjin Lu
- Department of Endocrinology & Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Xiaobin Li
- Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai 519000, Guangdong, China.
| | - Hongyun Lu
- Department of Endocrinology & Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China; Department of Endocrinology & Metabolism, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai 519000, Guangdong, China.
| |
Collapse
|
43
|
Hypoxia, Hypoxia-Inducible Factors and Liver Fibrosis. Cells 2021; 10:cells10071764. [PMID: 34359934 PMCID: PMC8305108 DOI: 10.3390/cells10071764] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/18/2022] Open
Abstract
Liver fibrosis is a potentially reversible pathophysiological event, leading to excess deposition of extracellular matrix (ECM) components and taking place as the net result of liver fibrogenesis, a dynamic and highly integrated process occurring during chronic liver injury of any etiology. Liver fibrogenesis and fibrosis, together with chronic inflammatory response, are primarily involved in the progression of chronic liver diseases (CLD). As is well known, a major role in fibrogenesis and fibrosis is played by activated myofibroblasts (MFs), as well as by macrophages and other hepatic cell populations involved in CLD progression. In the present review, we will focus the attention on the emerging pathogenic role of hypoxia, hypoxia-inducible factors (HIFs) and related mediators in the fibrogenic progression of CLD.
Collapse
|
44
|
Guo J, Akahoshi T, Mizuta Y, Murata M, Narahara S, Kawano T, Nagao Y, Zhang S, Tomikawa M, Kawanaka H, Hashizume M. Histidine-Rich Glycoprotein Alleviates Liver Ischemia/Reperfusion Injury in Mice With Nonalcoholic Steatohepatitis. Liver Transpl 2021; 27:840-853. [PMID: 33259137 DOI: 10.1002/lt.25960] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 09/28/2020] [Accepted: 11/03/2020] [Indexed: 12/30/2022]
Abstract
Hepatic ischemia/reperfusion injury (IRI) is a major complication of liver surgery and transplantation, especially in patients with nonalcoholic steatohepatitis (NASH). The mechanism of NASH susceptibility to IRI has not been fully clarified. We investigated the role of liver-produced histidine-rich glycoprotein (HRG) in NASH IRI. A NASH mouse model was established using C57BL/6J mice fed a methionine-choline-deficient diet (MCDD) for 6 weeks. The MCDD and standard diet groups were exposed to 60 minutes of partial hepatic ischemia/reperfusion (I/R). We further evaluated the impact of HRG in this context using HRG knockdown (KD) mice. IRI increased HRG expression in the standard diet group, but not in the MCDD group after I/R. HRG expression was inversely correlated with neutrophil infiltration and the formation of neutrophil extracellular traps (NETs). HRG KD mice showed severe liver injury with neutrophil infiltration and the formation of NETs. Pretreatment with supplementary HRG protected against I/R with the inhibition of neutrophil infiltration and the formation of NETs. In vitro, hepatocytes showed that the expression of HRG was upregulated under hypoxia/reoxygenation conditions, but not in response to oleic acid-treated hepatocytes. The decrease in HRG expression in fatty hepatocytes was accompanied by decreased farnesoid X receptor and hypoxia inducible factor 2 alpha subunit expression. HRG is a hepatoprotective factor during hepatic IRI because it decreases neutrophil infiltration and the formation of NETs. The decrease in HRG is a cause of susceptibility to IRI in steatotic livers. Therefore, HRG is a new therapeutic target for minimizing liver damage in patients with NASH.
Collapse
Affiliation(s)
- Jie Guo
- Department of Disaster and Emergency Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomohiko Akahoshi
- Department of Disaster and Emergency Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yukie Mizuta
- Department of Disaster and Emergency Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaharu Murata
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan
| | - Sayoko Narahara
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan
| | - Takahito Kawano
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Nagao
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan
| | - Shuo Zhang
- Department of Disaster and Emergency Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Morimasa Tomikawa
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan
| | - Hirofumi Kawanaka
- Clinical Research Institute, National Hospital Organization Beppu Medical Center, Beppu, Japan
| | - Makoto Hashizume
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
45
|
Mesarwi OA, Moya EA, Zhen X, Gautane M, Zhao H, Wegbrans Giró P, Alshebli M, McCarley KE, Breen EC, Malhotra A. Hepatocyte HIF-1 and Intermittent Hypoxia Independently Impact Liver Fibrosis in Murine NAFLD. Am J Respir Cell Mol Biol 2021; 65:390-402. [PMID: 34003729 DOI: 10.1165/rcmb.2020-0492oc] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Obstructive sleep apnea (OSA) is associated with insulin resistance, lipid dysregulation, and hepatic steatosis and fibrosis in nonalcoholic fatty liver disease (NAFLD). We have previously shown that hepatocyte hypoxia inducible factor-1 (HIF-1) mediates the development of liver fibrosis in a mouse model of NAFLD. We hypothesized that intermittent hypoxia (IH) modeling OSA would worsen hepatic steatosis and fibrosis in murine NAFLD, via HIF-1. Mice with hepatocyte-specific deletion of Hif1a (Hif1a-/-hep) and wild-type (Hif1aF/F) controls were fed a high trans-fat diet to induce NAFLD with steatohepatitis. Half from each group were exposed to IH, and the other half to intermittent air. Glucose tolerance test was performed prior to sacrifice. Liver collagen and triglycerides were determined. Mitochondrial efficiency was assessed in fresh liver tissue at sacrifice. Hepatic malondialdehyde concentration and pro-inflammatory cytokine levels were assessed, and genes of collagen and fatty acid metabolism were queried. Hif1a-/-hep mice gained less weight than Hif1aF/F mice (-2.3 grams, p=0.029). There was also a genotype-independent effect of IH on body weight, with less weight gain in IH (p=0.003). Fasting glucose, HOMA-IR, and glucose tolerance test were all improved in Hif1a-/-hep mice. Liver collagen was increased in IH (p=0.033), and reduced in Hif1a-/-hep mice (p<0.001), without any significant exposure/genotype interaction. Liver TNF-α and IL-1β were significantly increased in IH, and decreased in Hif1a-/-hep. We conclude that HIF-1 signaling worsens the metabolic profile and hastens NAFLD progression, and that IH may worsen liver fibrosis. These effects are plausibly mediated by hepatic inflammatory stress.
Collapse
Affiliation(s)
- Omar A Mesarwi
- University of California San Diego, 8784, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, California, United States;
| | - Esteban A Moya
- University of California San Diego, 8784, Division of Pulmonary, Critical Care, and Sleep Medicine, La Jolla, California, United States
| | - Xin Zhen
- University of California San Diego, 8784, La Jolla, California, United States
| | - Mary Gautane
- University of California San Diego, 8784, La Jolla, California, United States
| | - Huyai Zhao
- University of California San Diego, 8784, La Jolla, California, United States
| | - Paula Wegbrans Giró
- Imperial College London, 4615, Department of Bioengineering, London, United Kingdom of Great Britain and Northern Ireland
| | - Mouza Alshebli
- Abu Dhabi Health Services Co, 155078, Abu Dhabi, United Arab Emirates
| | - Kendall E McCarley
- University of Houston, 14743, Department of Educational Psychology, Houston, Texas, United States
| | - Ellen C Breen
- University of California San Diego, 8784, Division of Pulmonary, Critical Care, and Sleep Medicine, La Jolla, California, United States
| | - Atul Malhotra
- University of California San Diego, 8784, Division of Pulmonary, Critical Care, and Sleep Medicine, La Jolla, California, United States
| |
Collapse
|
46
|
Mooli RGR, Rodriguez J, Takahashi S, Solanki S, Gonzalez FJ, Ramakrishnan SK, Shah YM. Hypoxia via ERK Signaling Inhibits Hepatic PPARα to Promote Fatty Liver. Cell Mol Gastroenterol Hepatol 2021; 12:585-597. [PMID: 33798787 PMCID: PMC8258975 DOI: 10.1016/j.jcmgh.2021.03.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Fatty liver or nonalcoholic fatty liver disease (NAFLD) is the most common liver disease associated with comorbidities such as insulin resistance and cardiovascular and metabolic diseases. Chronic activation of hypoxic signaling, in particular, hypoxia-inducible factor (HIF)2α, promotes NAFLD progression by repressing genes involved in fatty acid β-oxidation through unclear mechanisms. Therefore, we assessed the precise mechanism by which HIF2α promotes fatty liver and its physiological relevance in metabolic homeostasis. METHODS Primary hepatocytes from VHL (VhlΔHep) and PPARα (Ppara-null) knockout mice that were loaded with fatty acids, murine dietary protocols to induce hepatic steatosis, and fasting-refeeding dietary regimen approaches were used to test our hypothesis. RESULTS Inhibiting autophagy using chloroquine did not decrease lipid contents in VhlΔHep primary hepatocytes. Inhibition of ERK using MEK inhibitor decreased lipid contents in primary hepatocytes from a genetic model of constitutive HIF activation and primary hepatocytes loaded with free fatty acids. Moreover, MEK-ERK inhibition potentiated ligand-dependent activation of PPARα. We also show that MEK-ERK inhibition improved diet-induced hepatic steatosis, which is associated with the induction of PPARα target genes. During fasting, fatty acid β-oxidation is induced by PPARα, and refeeding inhibits β-oxidation. Our data show that ERK is involved in the post-prandial repression of hepatic PPARα signaling. CONCLUSIONS Overall, our results demonstrate that ERK activated by hypoxia signaling plays a crucial role in fatty acid β-oxidation genes by repressing hepatocyte PPARα signaling.
Collapse
Affiliation(s)
- Raja Gopal Reddy Mooli
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jessica Rodriguez
- Department of Molecular and Integrative Physiology, Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Shogo Takahashi
- Departments of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia; National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sumeet Solanki
- Department of Molecular and Integrative Physiology, Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Frank J Gonzalez
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sadeesh K Ramakrishnan
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Molecular and Integrative Physiology, Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
47
|
How does hepatic lipid accumulation lead to lipotoxicity in non-alcoholic fatty liver disease? Hepatol Int 2021; 15:21-35. [PMID: 33548031 PMCID: PMC7886759 DOI: 10.1007/s12072-020-10121-2] [Citation(s) in RCA: 225] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD), characterized as excess lipid accumulation in the liver which is not due to alcohol use, has emerged as one of the major health problems around the world. The dysregulated lipid metabolism creates a lipotoxic environment which promotes the development of NAFLD, especially the progression from simple steatosis (NAFL) to non-alcoholic steatohepatitis (NASH). PURPOSEAND AIM This review focuses on the mechanisms of lipid accumulation in the liver, with an emphasis on the metabolic fate of free fatty acids (FFAs) in NAFLD and presents an update on the relevant cellular processes/mechanisms that are involved in lipotoxicity. The changes in the levels of various lipid species that result from the imbalance between lipolysis/lipid uptake/lipogenesis and lipid oxidation/secretion can cause organellar dysfunction, e.g. ER stress, mitochondrial dysfunction, lysosomal dysfunction, JNK activation, secretion of extracellular vesicles (EVs) and aggravate (or be exacerbated by) hypoxia which ultimately lead to cell death. The aim of this review is to provide an overview of how abnormal lipid metabolism leads to lipotoxicity and the cellular mechanisms of lipotoxicity in the context of NAFLD.
Collapse
|
48
|
Povero D, Johnson SM, Liu J. Hypoxia, hypoxia-inducible gene 2 (HIG2)/HILPDA, and intracellular lipolysis in cancer. Cancer Lett 2020; 493:71-79. [PMID: 32818550 PMCID: PMC11218043 DOI: 10.1016/j.canlet.2020.06.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/27/2020] [Accepted: 06/13/2020] [Indexed: 12/16/2022]
Abstract
Tumor tissues are chronically exposed to hypoxia owing to aberrant vascularity. Hypoxia induces metabolic alterations in cancer, thereby promoting aggressive malignancy and metastasis. While previous efforts largely focused on adaptive responses in glucose and glutamine metabolism, recent studies have begun to yield important insight into the hypoxic regulation of lipid metabolic reprogramming in cancer. Emerging evidence points to lipid droplet (LD) accumulation as a hallmark of hypoxic cancer cells. One critical underlying mechanism involves the inhibition of adipose triglyceride lipase (ATGL)-mediated intracellular lipolysis by a small protein encoded by hypoxia-inducible gene 2 (HIG2), also known as hypoxia inducible lipid droplet associated (HILPDA). In this review we summarize and discuss recent key findings on hypoxia-dependent regulation of metabolic adaptations especially lipolysis in cancer. We also pose several questions and hypotheses pertaining to the metabolic impact of lipolytic regulation in cancer under hypoxia and during hypoxia-reoxygenation transition.
Collapse
Affiliation(s)
- Davide Povero
- From Department of Biochemistry and Molecular Biology, Rochester, MN, 55905, USA; Division of Endocrinology, Rochester, MN, 55905, USA
| | - Scott M Johnson
- From Department of Biochemistry and Molecular Biology, Rochester, MN, 55905, USA; Mayo Clinic College of Medicine & Science, Rochester, MN, 55905, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, 55905, USA
| | - Jun Liu
- From Department of Biochemistry and Molecular Biology, Rochester, MN, 55905, USA; Division of Endocrinology, Rochester, MN, 55905, USA.
| |
Collapse
|
49
|
Zhang X, Huang C, Li X, Shangguan Z, Wei W, Liu S, Yang S, Liu Y. HFD and HFD-provoked hepatic hypoxia act as reciprocal causation for NAFLD via HIF-independent signaling. BMC Gastroenterol 2020; 20:366. [PMID: 33143650 PMCID: PMC7640429 DOI: 10.1186/s12876-020-01515-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/26/2020] [Indexed: 12/30/2022] Open
Abstract
Background The occurrence of non-alcoholic fatty liver disease (NAFLD) is found to be higher in patients with obstructive sleep apnea (OSA), which is characterized by intermittent hypoxia. Activation of hypoxia-inducible factors has been shown in the development and progression of NAFLD, implying a cause and effects relationship between NAFLD and hypoxia. The present study was designed to investigate the interaction of lipotoxicity and hypoxia in the pathogenesis of NAFLD using mice model with high-fat diet (HFD) feeding or hypoxic treatment. Methods NAFLD model was induced in mice by HFD feeding, and in cultured primary hepatocytes by administration of palmitate acid. Mouse hypoxic model was produced by placing the mice in a Animal incubator with oxygen concentration at 75% followed by a 21% oxygen supplement. Hypoxic condition was mimicked by treating the hepatocytes with cobalt chloride (CoCl2) or 1% oxygen supply. Pimonidazole assay was conducted to evaluate hypoxia. Lipid metabolic genes were measured by real-time polymerase-chain reaction. HIF-1α and HIF-2α genes were silenced by siRNA. Results HFD feeding and palmitate acid treatment provoked severe hepatic hypoxia along with TG accumulation in mice and in cultured primary hepatocytes respectively. Conversely, hypoxia induced hepatic TG accumulation in mice and in cultured primary hepatocytes. Hypoxic treatment inhibited the expression of lipolytic genes, while increased the expression of lipogenicgenes in mice. Although both lipotoxicity and hypoxia could activate hepatic hypoxia-induced factor 1α and 2α, while neither lipotoxicity- nor hypoxia- induced hepatic steatosis was affected when HIF was knocked down. Conclusions HFD resulted in hepatic TG accumulation and concomitant hypoxia. Conversely, hypoxia induced hepatic TG accumulation in mice and in cultured heptocytes. Thus lipotoxicity and hypoxia might work as reciprocal causation and orchestrate to promote the development of NAFLD.
Collapse
Affiliation(s)
- Xiaofang Zhang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Road, Xiamen, 361003, China.,Xiamen Clinical Medical Center for Endocrine and Metabolic Diseases, Xiamen, China.,Fujian Province Key Laboratory of Diabetes Translational Medicine, Xiamen, China
| | - Caoxin Huang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Road, Xiamen, 361003, China.,Xiamen Clinical Medical Center for Endocrine and Metabolic Diseases, Xiamen, China.,Fujian Province Key Laboratory of Diabetes Translational Medicine, Xiamen, China
| | - Xuejun Li
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Road, Xiamen, 361003, China.,Xiamen Clinical Medical Center for Endocrine and Metabolic Diseases, Xiamen, China.,Fujian Province Key Laboratory of Diabetes Translational Medicine, Xiamen, China.,Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Zhaoshui Shangguan
- Research Center for Translational Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Wenjing Wei
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Road, Xiamen, 361003, China.,Xiamen Clinical Medical Center for Endocrine and Metabolic Diseases, Xiamen, China.,Fujian Province Key Laboratory of Diabetes Translational Medicine, Xiamen, China
| | - Suhuan Liu
- Research Center for Translational Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Shuyu Yang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Road, Xiamen, 361003, China. .,Xiamen Clinical Medical Center for Endocrine and Metabolic Diseases, Xiamen, China. .,Fujian Province Key Laboratory of Diabetes Translational Medicine, Xiamen, China.
| | - Yijie Liu
- Siming Branch, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Road, Xiamen, 361003, China.
| |
Collapse
|
50
|
Li T, Mao C, Wang X, Shi Y, Tao Y. Epigenetic crosstalk between hypoxia and tumor driven by HIF regulation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:224. [PMID: 33109235 PMCID: PMC7592369 DOI: 10.1186/s13046-020-01733-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
Hypoxia is the major influence factor in physiological and pathological courses which are mainly mediated by hypoxia-inducible factors (HIFs) in response to low oxygen tensions within solid tumors. Under normoxia, HIF signaling pathway is inhibited due to HIF-α subunits degradation. However, in hypoxic conditions, HIF-α is activated and stabilized, and HIF target genes are successively activated, resulting in a series of tumour-specific activities. The activation of HIFs, including HIF-1α, HIF-2α and HIF-3α, subsequently induce downstream target genes which leads to series of responses, the resulting abnormal processes or metabolites in turn affect HIFs stability. Given its functions in tumors progression, HIFs have been regarded as therapeutic targets for improved treatment efficacy. Epigenetics refers to alterations in gene expression that are stable between cell divisions, and sometimes between generations, but do not involve changes in the underlying DNA sequence of the organism. And with the development of research, epigenetic regulation has been found to play an important role in the development of tumors, which providing accumulating basic or clinical evidences for tumor treatments. Here, given how little has been reported about the overall association between hypoxic tumors and epigenetics, we made a more systematic review from epigenetic perspective in hope of helping others better understand hypoxia or HIF pathway, and providing more established and potential therapeutic strategies in tumors to facilitate epigenetic studies of tumors.
Collapse
Affiliation(s)
- Tiansheng Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Chao Mao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiang Wang
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Ying Shi
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| | - Yongguang Tao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China. .,Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|