1
|
Zhang N, Shen S, Yang M, He S, Liu C, Li H, Lu T, Liu H, Hu Q, Tang W, Chen Y. Design, Synthesis, and Biological Evaluation of a Novel NIK Inhibitor with Anti-Inflammatory and Hepatoprotective Effects for Sepsis Treatment. J Med Chem 2024; 67:5617-5641. [PMID: 38563549 DOI: 10.1021/acs.jmedchem.3c02266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
NIK plays a crucial role in the noncanonical NF-κB signaling pathway associated with diverse inflammatory and autoimmune diseases. Our study presents compound 54, a novel NIK inhibitor, designed through a structure-based scaffold-hopping approach from the previously identified B022. Compound 54 demonstrates remarkable selectivity and potency against NIK both in vitro and in vivo, effectively suppressing pro-inflammatory cytokines and nitric oxide production. In mouse models, compound 54 protected against LPS-induced systemic sepsis, reducing AST, ALT, and AKP liver injury markers. Additionally, it also attenuates sepsis-induced lung and kidney damage. Mechanistically, compound 54 blocks the noncanonical NF-κB signaling pathway by targeting NIK, preventing p100 to p52 processing. This work reveals a novel class of NIK inhibitors with significant potential for sepsis therapy.
Collapse
Affiliation(s)
- Nanxia Zhang
- School of Sciences, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Shige Shen
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Mengyu Yang
- School of Sciences, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Sijie He
- School of Sciences, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Chunxiao Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Hongmei Li
- School of Sciences, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Tao Lu
- School of Sciences, China Pharmaceutical University, Nanjing 211198, P. R. China
- State Key Laboratory of Natural Medicines, School of Science, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Haichun Liu
- Laboratory of Molecular Design and Drug Discovery, School of Sciences, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Qinghua Hu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Weifang Tang
- School of Sciences, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Yadong Chen
- School of Sciences, China Pharmaceutical University, Nanjing 211198, P. R. China
- State Key Laboratory of Natural Medicines, School of Science, China Pharmaceutical University, Nanjing 211198, P. R. China
| |
Collapse
|
2
|
Tang Y. Analysis of the binding pattern of NIK inhibitors by computational simulation. J Biomol Struct Dyn 2024; 42:3318-3331. [PMID: 37183664 DOI: 10.1080/07391102.2023.2212782] [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: 10/29/2022] [Accepted: 05/04/2023] [Indexed: 05/16/2023]
Abstract
NF-kappaB-Inducing Kinase (NIK) is a key kinase in the activation of the NF-κB non-classical signalling pathway, which has been shown to be over-activated in patients with inflammatory diseases, immune disorders and malignancies and solid tumours inducing activation of the NF-κB non-classical signalling pathway. The design of ATP-competitive small molecule inhibitors against NIK has been a hot topic in the last decade, and many efficient NIK inhibitors have been identified. In this work, I aim to unravel the mechanism of NIK inhibition by different representative NIK type I 1/2 kinase inhibitors, using ADME, molecular docking, molecular dynamics simulation, MM-PBSA analysis and 3D-QSAR analysis. This work contributes to the understanding of the efficiency of NIK inhibitor binding by revealing the basis of the efficiency of NIK inhibitors, the difference in binding modes between different inhibitors and the overall effect on NIK.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yingkai Tang
- Department of Anatomy, School of Basic Medicine, Bengbu Medical University, Bengbu, China
| |
Collapse
|
3
|
Lin Y, Liu S, Sun Y, Chen C, Yang S, Pei G, Lin M, Yu J, Liu X, Wang H, Long J, Yan Q, Liang J, Yao J, Yi F, Meng L, Tan Y, Chen N, Yang Y, Ai Q. CCR5 and inflammatory storm. Ageing Res Rev 2024; 96:102286. [PMID: 38561044 DOI: 10.1016/j.arr.2024.102286] [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: 12/28/2023] [Revised: 03/15/2024] [Accepted: 03/25/2024] [Indexed: 04/04/2024]
Abstract
Chemokines and their corresponding receptors play crucial roles in orchestrating inflammatory and immune responses, particularly in the context of pathological conditions disrupting the internal environment. Among these receptors, CCR5 has garnered considerable attention due to its significant involvement in the inflammatory cascade, serving as a pivotal mediator of neuroinflammation and other inflammatory pathways associated with various diseases. However, a notable gap persists in comprehending the intricate mechanisms governing the interplay between CCR5 and its ligands across diverse and intricate inflammatory pathologies. Further exploration is warranted, especially concerning the inflammatory cascade instigated by immune cell infiltration and the precise binding sites within signaling pathways. This study aims to illuminate the regulatory axes modulating signaling pathways in inflammatory cells by providing a comprehensive overview of the pathogenic processes associated with CCR5 and its ligands across various disorders. The primary focus lies on investigating the pathomechanisms associated with CCR5 in disorders related to neuroinflammation, alongside the potential impact of aging on these processes and therapeutic interventions. The discourse culminates in addressing current challenges and envisaging potential future applications, advocating for innovative research endeavors to advance our comprehension of this realm.
Collapse
Affiliation(s)
- Yuting Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shasha Liu
- Department of Pharmacy, Changsha Hospital for Matemal&Child Health Care Affiliated to Hunan Normal University, Changsha 410007, China
| | - Yang Sun
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Chen Chen
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Songwei Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Gang Pei
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Meiyu Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jingbo Yu
- Technology Innovation Center/National Key Laboratory Breeding Base of Chinese Medicine Powders and Innovative Drugs, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xuan Liu
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Huiqin Wang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Junpeng Long
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Qian Yan
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jinping Liang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jiao Yao
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Fan Yi
- Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing 100048, China
| | - Lei Meng
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yong Tan
- Nephrology Department, Xiangtan Central Hospital, Xiangtan 411100, China
| | - Naihong Chen
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yantao Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| | - Qidi Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| |
Collapse
|
4
|
Wang P, Kang Q, Wu WS, Rui L. Hepatic Snai1 and Snai2 promote liver regeneration and suppress liver fibrosis in mice. Cell Rep 2024; 43:113875. [PMID: 38451818 PMCID: PMC11025633 DOI: 10.1016/j.celrep.2024.113875] [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: 11/21/2022] [Revised: 12/21/2023] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
Liver injury stimulates hepatocyte replication and hepatic stellate cell (HSC) activation, thereby driving liver regeneration. Aberrant HSC activation induces liver fibrosis. However, mechanisms underlying liver regeneration and fibrosis remain poorly understood. Here, we identify hepatic Snai1 and Snai2 as important transcriptional regulators for liver regeneration and fibrosis. Partial hepatectomy or CCl4 treatment increases occupancies of Snai1 and Snai2 on cyclin A2 and D1 promoters in the liver. Snai1 and Snai2 in turn increase promoter H3K27 acetylation and cyclin A2/D1 expressions. Hepatocyte-specific deletion of both Snai1 and Snai2, but not one alone, suppresses liver cyclin A2/D1 expression and regenerative hepatocyte proliferation after hepatectomy or CCl4 treatments but augments CCl4-stimulated HSC activation and liver fibrosis. Conversely, Snai2 overexpression in the liver enhances hepatocyte replication and suppresses liver fibrosis after CCl4 treatment. These results suggest that hepatic Snai1 and Snai2 directly promote, via histone modifications, reparative hepatocyte replication and indirectly inhibit liver fibrosis.
Collapse
Affiliation(s)
- Pingping Wang
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; School of Food Science and Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510640, China; School of Chemical Engineering and Light Insulation, Guangdong University of Technology, Guangzhou 510006, China
| | - Qianqian Kang
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Wen-Shu Wu
- Division of Hematology/Oncology, Department of Medicine, UI Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Liangyou Rui
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Elizabeth Weiser Caswell Diabetes Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
5
|
Roy D, Chatterjee A, Mishra L, Chakraborty N. Progression of retinal choroidal neovascularization by latent human cytomegalovirus infection and immunological signaling among neonatal patients admitted to tertiary care hospital. J Med Virol 2024; 96:e29478. [PMID: 38377063 DOI: 10.1002/jmv.29478] [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: 12/29/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/22/2024]
Abstract
Choroidal neovascularization (CNV) is a serious condition that affects the retina, causing partial or complete blindness in people of different ages. While CNV is a common occurrence in various chorioretinopathies, research on its occurrence in neonates is limited. Human cytomegalovirus (HCMV) is a significant health threat to neonates, with a strong association with retinal angiogenesis. However, there has been limited investigation into HCMV-associated CNV progression. In this article, we extensively studied the expression of different inflammatory cytokines and chemokines during latent HCMV-associated retinal neovascularization. Our research found that HCMV-induced CNV progression was significantly prominent in the presence of AT2R-dependent angiogenesis (p < 0.001), whereas in the absence of HCMV, AT1R-dependent CCL-5-mediated angiogenesis was documented. We also observed significant increases in CCL-19, CCL-21 chemokine responses, followed by CCR-7 chemokine receptor activation (p < 0.001) in HCMV-induced CNV patients compared to HCMV non-induced CNV groups. Furthermore, significant changes in predictive chemokine markers of HCMV-induced CNV were positively correlated with HCMV viremia. These immunological alterations ultimately lead to the switching of NFκB canonical and noncanonical pathways, respectively, in HCMV-induced neonatal CNV and HCMV non-induced CNV. This clinical observation presents a novel hypothesis that ocular HCMV latency poses a noteworthy risk factor for the progression of retinal neovascularization through a distinctive immunological signaling pathway. The current study represents the first of its kind to report on this association, which may have significant implications for the clinical management of patients with ocular HCMV.
Collapse
Affiliation(s)
- Debsopan Roy
- Virus Research Laboratory, ICMR-NICED, Kolkata, West Bengal, India
| | - Aroni Chatterjee
- Department of Biotechnology, School of Biotechnology and Bioscience, Brainware University, Kolkata, West Bengal, India
| | - Lopamudra Mishra
- Department of Pediatrics, IPGME&R, SSKM Hospital, Kolkata, West Bengal, India
| | | |
Collapse
|
6
|
Xu C, Zhou H, Jin Y, Sahay K, Robicsek A, Liu Y, Dong K, Zhou J, Barrett A, Su H, Chen W. Hepatic neddylation deficiency triggers fatal liver injury via inducing NF-κB-inducing kinase in mice. Nat Commun 2022; 13:7782. [PMID: 36526632 PMCID: PMC9758150 DOI: 10.1038/s41467-022-35525-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
The conjugation of neural precursor cell expressed, developmentally downregulated 8 (NEDD8) to target proteins, termed neddylation, participates in many cellular processes and is aberrant in various pathological diseases. Its relevance to liver function and failure remains poorly understood. Herein, we show dysregulated expression of NAE1, a regulatory subunit of the only NEDD8 E1 enzyme, in human acute liver failure. Embryonic- and adult-onset deletion of NAE1 in hepatocytes causes hepatocyte death, inflammation, and fibrosis, culminating in fatal liver injury in mice. Hepatic neddylation deficiency triggers oxidative stress, mitochondrial dysfunction, and hepatocyte reprogramming, potentiating liver injury. Importantly, NF-κB-inducing kinase (NIK), a serine/Thr kinase, is a neddylation substrate. Neddylation of NIK promotes its ubiquitination and degradation. Inhibition of neddylation conversely causes aberrant NIK activation, accentuating hepatocyte damage and inflammation. Administration of N-acetylcysteine, a glutathione surrogate and antioxidant, mitigates liver failure caused by hepatic NAE1 deletion in adult male mice. Therefore, hepatic neddylation is important in maintaining postnatal and adult liver homeostasis, and the identified neddylation targets/pathways provide insights into therapeutically intervening acute liver failure.
Collapse
Affiliation(s)
- Cheng Xu
- grid.410427.40000 0001 2284 9329Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Hongyi Zhou
- grid.410427.40000 0001 2284 9329Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Yulan Jin
- grid.410427.40000 0001 2284 9329Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Khushboo Sahay
- grid.410427.40000 0001 2284 9329Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Anna Robicsek
- grid.410427.40000 0001 2284 9329Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Yisong Liu
- grid.410427.40000 0001 2284 9329Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Kunzhe Dong
- grid.410427.40000 0001 2284 9329Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Jiliang Zhou
- grid.410427.40000 0001 2284 9329Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Amanda Barrett
- grid.410427.40000 0001 2284 9329Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Huabo Su
- grid.410427.40000 0001 2284 9329Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Weiqin Chen
- grid.410427.40000 0001 2284 9329Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| |
Collapse
|
7
|
Ishtiaq SM, Arshad MI, Khan JA. PPARγ signaling in hepatocarcinogenesis: Mechanistic insights for cellular reprogramming and therapeutic implications. Pharmacol Ther 2022; 240:108298. [PMID: 36243148 DOI: 10.1016/j.pharmthera.2022.108298] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 11/30/2022]
Abstract
Liver cancer or hepatocellular carcinoma (HCC) is leading cause of cancer-related mortalities globally. The therapeutic approaches for chronic liver diseases-associated liver cancers aimed at modulating immune check-points and peroxisome proliferator-activated receptor gamma (PPARγ) signaling pathway during multistep process of hepatocarcinogenesis that played a dispensable role in immunopathogenesis and outcomes of disease. Herein, the review highlights PPARγ-induced effects in balancing inflammatory (tumor necrosis factor alpha (TNF-α), interleukin (IL)-6, IL-1) and anti-inflammatory cytokines (IL-10, transforming growth factor beta (TGF-β), and interplay of PPARγ, hepatic stellate cells and fibrogenic niche in cell-intrinsic and -extrinsic crosstalk of hepatocarcinogenesis. PPARγ-mediated effects in pre-malignant microenvironment promote growth arrest, cell senescence and cell clearance in liver cancer pathophysiology. Furthermore, PPARγ-immune cell axis of liver microenvironment exhibits an immunomodulation strategy of resident immune cells of the liver (macrophages, natural killer cells, and dendritic cells) in concomitance with current clinical guidelines of the European Association for Study of Liver Diseases (EASL) for several liver diseases. Thus, mechanistic insights of PPARγ-associated high value targets and canonical signaling suggest PPARγ as a possible therapeutic target in reprogramming of hepatocarcinogenesis to decrease burden of liver cancers, worldwide.
Collapse
Affiliation(s)
- Syeda Momna Ishtiaq
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad 38040, Pakistan
| | | | - Junaid Ali Khan
- Department of Pharmacology and Physiology, MNS University of Agriculture, Multan 60000, Pakistan.
| |
Collapse
|
8
|
Angelica sinensis Polysaccharide and Astragalus membranaceus Polysaccharide Accelerate Liver Regeneration by Enhanced Glycolysis via Activation of JAK2/STAT3/HK2 Pathway. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227890. [PMID: 36431990 PMCID: PMC9695464 DOI: 10.3390/molecules27227890] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/05/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022]
Abstract
The promotion of liver regeneration is crucial to avoid liver failure after hepatectomy. Angelica sinensis polysaccharide (ASP) and Astragalus membranaceus polysaccharide (AMP) have been identified as being associated with hepatoprotective effects. However, their roles and specific mechanisms in liver regeneration remain to be elucidated. In the present study, it suggested that the respective use of ASP or AMP strikingly promoted hepatocyte proliferation in vitro with a wide range of concentrations (from 12.5 μg/mL to 3200 μg/mL), and a stronger promoting effect was observed in combined interventions. A significantly enhanced liver/body weight ratio (4.20%) on day 7 and reduced serum transaminase (ALT 243.53 IU/L and AST 423.74 IU/L) and total bilirubin (52.61 IU/L) levels on day 3 were achieved by means of ASP-AMP administration after partial hepatectomy in mice. Metabonomics showed that differential metabolites were enriched in glycolysis with high expression of beta-d-fructose 6-phosphate and lactate, followed by significantly strengthened lactate secretion in the supernatant (0.54) and serum (0.43) normalized to control. Upon ASP-AMP treatment, the knockdown of hexokinase 2 (HK2) or inhibited glycolysis caused by 2-deoxy-d-glucose decreased hepatocyte proliferation in vitro and in vivo. Furthermore, pathway analysis predicted the role of JAK2/STAT3 pathway in ASP-AMP-regulated liver regeneration, and phosphorylation of JAK2 and STAT3 was proven to be elevated in this promoting process. Finally, downregulated expression of HK2, an attenuated level of lactate secretion, and reduced hepatocyte proliferation were displayed when STAT3 was knocked out in vitro. Therefore, it can be concluded that ASP-AMP accelerated liver regeneration and exerted a hepatoprotective effect after hepatectomy, in which the JAK2/STAT3/HK2 pathway was actively involved in activating glycolysis.
Collapse
|
9
|
Vesting AJ, Jais A, Klemm P, Steuernagel L, Wienand P, Fog-Tonnesen M, Hvid H, Schumacher AL, Kukat C, Nolte H, Georgomanolis T, Altmüller J, Pasparakis M, Schmidt A, Krüger M, Supprian MS, Waisman A, Straub BK, Raschzok N, Bernier M, Birkenfeld AL, Hövelmeyer N, Brüning JC, Wunderlich FT. NIK/MAP3K14 in hepatocytes orchestrates NASH to hepatocellular carcinoma progression via JAK2/STAT5 inhibition. Mol Metab 2022; 66:101626. [PMID: 36356831 PMCID: PMC9676392 DOI: 10.1016/j.molmet.2022.101626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/21/2022] [Accepted: 10/28/2022] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVE Nonalcoholic fatty liver disease (NAFLD) ranges from steatosis to nonalcoholic steatohepatitis (NASH), which often progresses to hepatocellular carcinoma (HCC) through a largely undefined mechanism. NASH and HCC depend on inflammatory signaling, whose master regulator is the NFκB transcription factor family, activated by canonical and non-canonical pathways. METHODS Here, we investigated non-canonical NFκB-inducing kinase (NIK/MAP3K14) in metabolic NASH, NASH to HCC transition, and DEN-induced HCC. To this end, we performed dietary and chemical interventions in mice that were analyzed via single nucleus sequencing, gene expression and histochemical methods. Ultimately, we verified our mouse results in human patient samples. RESULTS We revealed that hepatocyte-specific NIK deficiency (NIKLKO) ameliorated metabolic NASH complications and reduced hepatocarcinogenesis, independent of its role in the NFκB pathway. Instead, hepatic NIK attenuated hepatoprotective JAK2/STAT5 signaling that is a prerequisite for NASH and NASH to HCC progression in mice and humans. CONCLUSIONS Our data suggest NIK-mediated inhibitory JAK2 phosphorylation at serine 633 that might be amenable for future therapeutic interventions in patients.
Collapse
Affiliation(s)
- Anna Juliane Vesting
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany, Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany, Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Alexander Jais
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany, Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany, Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), 04103 Leipzig, Germany
| | - Paul Klemm
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany, Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany, Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Lukas Steuernagel
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany, Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany, Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Peter Wienand
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany, Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany, Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Morten Fog-Tonnesen
- Global Drug Discovery, Novo Nordisk A/S, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| | - Henning Hvid
- Pathology & Imaging, Novo Nordisk A/S, Novo Nordisk Park 1, DK-2760 Maaloev, Denmark
| | - Anna-Lena Schumacher
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, 50931 Cologne, Germany
| | - Christian Kukat
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, 50931 Cologne, Germany
| | - Hendrik Nolte
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, 50931 Cologne, Germany
| | | | - Janine Altmüller
- University of Cologne, Cologne Center for Genomics, Cologne, Germany
| | - Manolis Pasparakis
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany, Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Andreas Schmidt
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany, Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Marcus Krüger
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany, Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Marc Schmidt Supprian
- Institute of Experimental Hematology, TranslaTUM, Klinikum rechts der Isar der Technischen Universität München, 81675 Munich, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) 69120 Heidelberg, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Beate Katharina Straub
- Institute of Pathology, University Medical Centre of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Nathanael Raschzok
- General, Visceral, and Transplantation Surgery, Charité-University School of Medicine, 13353 Berlin, Germany- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Surgery, Experimental Surgery, Campus Charité Mitte | Campus Virchow-Klinikum, Berlin, Germany and Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Academy, Clinician Scientist Program, Berlin, Germany
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Andreas L Birkenfeld
- Internal Medicine IV, Clinic of Diabetology, Endocrinology, Nephrology, Internal medicine IV, University Hospital and Faculty of Medicine of the Eberhard Karls University Tübingen, 72016 Tübingen, Germany and Institute of Diabetes Research and Metabolic Diseases, Helmholtz Zentrum München an der Uniklinik Tübingen, Deutsches Zentrum für Diabetesforschung (DZD), Germany
| | - Nadine Hövelmeyer
- Institute for Molecular Medicine, Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany, Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany, Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - F Thomas Wunderlich
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany, Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany, Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
10
|
Biliary NIK promotes ductular reaction and liver injury and fibrosis in mice. Nat Commun 2022; 13:5111. [PMID: 36042192 PMCID: PMC9427946 DOI: 10.1038/s41467-022-32575-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 08/08/2022] [Indexed: 11/21/2022] Open
Abstract
Excessive cholangiocyte expansion (ductular reaction) promotes liver disease progression, but the underlying mechanism is poorly understood. Here we identify biliary NF-κB-inducing kinase (NIK) as a pivotal regulator of ductular reaction. NIK is known to activate the noncanonical IKKα/NF-κB2 pathway and regulate lymphoid tissue development. We find that cholangiocyte NIK is upregulated in mice with cholestasis induced by bile duct ligation (BDL), 5-diethoxycarbonyl-1,4-dihydrocollidine (DDC), or α-naphtyl-isothiocyanate (ANIT). DDC, ANIT, or BDL induces ductular reaction, liver injury, inflammation, and fibrosis in mice. Cholangiocyte-specific deletion of NIK, but not IKKα, blunts these pathological alterations. NIK inhibitor treatment similarly ameliorates DDC-induced ductular reaction, liver injury, and fibrosis. Biliary NIK directly increases cholangiocyte proliferation while suppressing cholangiocyte death, and it also promotes secretion of cholangiokines from cholangiocytes. Cholangiokines stimulate liver macrophages and hepatic stellate cells, augmenting liver inflammation and fibrosis. These results unveil a NIK/ductular reaction axis and a NIK/cholangiokine axis that promote liver disease progression. Excessive expansion of cholangiocytes in the liver leads to ductular reaction and liver disease. Here, the authors show that genetic ablation, or pharmacological inhibition, of biliary NIK blocks ductular reaction, liver inflammation, and liver fibrosis in mice by modulating secretion of cholangiokines that mediate liver inflammation and fibrosis.
Collapse
|
11
|
Tao L, Ren X, Zhai W, Chen Z. Progress and Prospects of Non-Canonical NF-κB Signaling Pathway in the Regulation of Liver Diseases. Molecules 2022; 27:molecules27134275. [PMID: 35807520 PMCID: PMC9268066 DOI: 10.3390/molecules27134275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/04/2023] Open
Abstract
Non-canonical nuclear factor kappa B (NF-κB) signaling pathway regulates many physiological and pathological processes, including liver homeostasis and diseases. Recent studies demonstrate that non-canonical NF-κB signaling pathway plays an essential role in hyperglycemia, non-alcoholic fatty liver disease, alcoholic liver disease, liver regeneration, liver injury, autoimmune liver disease, viral hepatitis, and hepatocellular carcinoma. Small-molecule inhibitors targeting to non-canonical NF-κB signaling pathway have been developed and shown promising results in the treatment of liver injuries. Here, the recent advances and future prospects in understanding the roles of the non-canonical NF-κB signaling pathways in the regulation of liver diseases are discussed.
Collapse
Affiliation(s)
- Li Tao
- Emergency Department, 305 Hospital of People’s Liberation Army, Beijing 100017, China; (L.T.); (W.Z.)
| | - Xiaomeng Ren
- College of Pharmaceutical and Biology Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
- Correspondence: (X.R.); (Z.C.); Tel.: +86-45186402029 (Z.C.)
| | - Wenhui Zhai
- Emergency Department, 305 Hospital of People’s Liberation Army, Beijing 100017, China; (L.T.); (W.Z.)
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
- Correspondence: (X.R.); (Z.C.); Tel.: +86-45186402029 (Z.C.)
| |
Collapse
|
12
|
Fan Z, Kong M, Dong W, Dong C, Miao X, Guo Y, Liu X, Miao S, Li L, Chen T, Qu Y, Yu F, Duan Y, Lu Y, Zou X. Trans-activation of eotaxin-1 by Brg1 contributes to liver regeneration. Cell Death Dis 2022; 13:495. [PMID: 35614068 PMCID: PMC9132924 DOI: 10.1038/s41419-022-04944-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 12/14/2022]
Abstract
Infiltration of eosinophils is associated with and contributes to liver regeneration. Chemotaxis of eosinophils is orchestrated by the eotaxin family of chemoattractants. We report here that expression of eotaxin-1 (referred to as eotaxin hereafter), but not that of either eotaxin-2 or eotaxin-3, were elevated, as measured by quantitative PCR and ELISA, in the proliferating murine livers compared to the quiescent livers. Similarly, exposure of primary murine hepatocytes to hepatocyte growth factor (HGF) stimulated eotaxin expression. Liver specific deletion of Brahma-related gene 1 (Brg1), a chromatin remodeling protein, attenuated eosinophil infiltration and down-regulated eotaxin expression in mice. Brg1 deficiency also blocked HGF-induced eotaxin expression in cultured hepatocytes. Further analysis revealed that Brg1 could directly bind to the proximal eotaxin promoter to activate its transcription. Mechanistically, Brg1 interacted with nuclear factor kappa B (NF-κB)/RelA to activate eotaxin transcription. NF-κB knockdown or pharmaceutical inhibition disrupted Brg1 recruitment to the eotaxin promoter and blocked eotaxin induction in hepatocytes. Adenoviral mediated over-expression of eotaxin overcame Brg1 deficiency caused delay in liver regeneration in mice. On the contrary, eotaxin depletion with RNAi or neutralizing antibodies retarded liver regeneration in mice. More important, Brg1 expression was detected to be correlated with eotaxin expression and eosinophil infiltration in human liver specimens. In conclusion, our data unveil a novel role of Brg1 as a regulator of eosinophil trafficking by activating eotaxin transcription.
Collapse
Affiliation(s)
- Zhiwen Fan
- grid.428392.60000 0004 1800 1685Department of Pathology, Nanjing Drum Tower Hospital Affiliated with Nanjing University School of Medicine, Nanjing, China ,grid.428392.60000 0004 1800 1685Department of Gastroenterology, Nanjing Drum Tower Hospital Affiliated with Nanjing University School of Medicine, Nanjing, China
| | - Ming Kong
- grid.89957.3a0000 0000 9255 8984Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Wenhui Dong
- grid.89957.3a0000 0000 9255 8984Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Chunlong Dong
- grid.410745.30000 0004 1765 1045Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiulian Miao
- grid.411351.30000 0001 1119 5892College of Life Sciences and Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Yan Guo
- grid.411351.30000 0001 1119 5892College of Life Sciences and Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Xingyu Liu
- grid.411351.30000 0001 1119 5892College of Life Sciences and Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Shuying Miao
- grid.428392.60000 0004 1800 1685Department of Pathology, Nanjing Drum Tower Hospital Affiliated with Nanjing University School of Medicine, Nanjing, China
| | - Lin Li
- grid.428392.60000 0004 1800 1685Department of Pathology, Nanjing Drum Tower Hospital Affiliated with Nanjing University School of Medicine, Nanjing, China
| | - Tingting Chen
- grid.428392.60000 0004 1800 1685Department of Pathology, Nanjing Drum Tower Hospital Affiliated with Nanjing University School of Medicine, Nanjing, China
| | - Yeqing Qu
- grid.428392.60000 0004 1800 1685Experimental Animal Center, Nanjing Drum Tower Hospital Affiliated with Nanjing University School of Medicine, Nanjing, China
| | - Fei Yu
- grid.428392.60000 0004 1800 1685Experimental Animal Center, Nanjing Drum Tower Hospital Affiliated with Nanjing University School of Medicine, Nanjing, China
| | - Yunfei Duan
- grid.490563.d0000000417578685Department of Hepatobiliary Surgery, the First People’s Hospital of Changzhou, the Third Hospital Affiliated with Soochow University, Changzhou, China
| | - Yunjie Lu
- grid.490563.d0000000417578685Department of Hepatobiliary Surgery, the First People’s Hospital of Changzhou, the Third Hospital Affiliated with Soochow University, Changzhou, China
| | - Xiaoping Zou
- grid.428392.60000 0004 1800 1685Department of Gastroenterology, Nanjing Drum Tower Hospital Affiliated with Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
13
|
Popov SS, Kryl'skii ED, Shulgin KK, Raskina EA, Popova TN, Pashkov AN, Kuptsova GN. Inflammation is associated with impairment of oxidative status, carbohydrate and lipid metabolism in type 2 diabetes complicated by non-alcoholic fatty liver disease. Minerva Endocrinol (Torino) 2022; 47:304-313. [PMID: 35195377 DOI: 10.23736/s2724-6507.20.03257-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is accompanied by inflammation and impairment of the lipid metabolism. In addition, NAFLD is one of the major complications of type 2 diabetes associated with oxidative stress. Based on this, we evaluated the tumour necrosis factor alpha (TNF-α), nuclear factor κB (NF-κB), oxidative status rates, and analysed its correlation with carbohydrate and lipid metabolism in patients with NAFLD and type 2 diabetes. METHODS A case-control study included 63 participants with NAFLD developing in patients with type 2 diabetes, and 65 healthy volunteers with a normal complete blood count and blood biochemical profile. The following parameters and states were assessed during the study: glycaemia, insulin resistance, lipid levels, liver tests, intensity of free radical induced oxidation, antioxidant enzymes, TNF-α and NF-κB level. RESULTS Free radical induced oxidation was significantly elevated (p<0.001), total antioxidant activity was significantly decreased (p<0.001) and associated with insulin resistance (p=0.019) and lipid metabolism shifts (p<0.05) in patients with NAFLD and type 2 diabetes. Such patients had showed impaired functioning of antioxidant system (p<0.001), inhibition of NADPH-generating enzymes activity (p<0.001), increased levels of TNF-α (p<0.001) and NF-κB (p=0.019) correlated with the severity of hyperglycaemia (p<0.05), concentration of reduced glutathione (p=0.005) and total cholesterol (p=0.016). CONCLUSIONS The increase of free radical induced oxidation, TNF-α and NF-κB levels, and depletion of the antioxidant system seems to be the key factors of the development of NAFLD in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Sergey S Popov
- Department of Hospital therapy and endocrinology, Voronezh State Medical University named after N.N. Burdenko, Voronezh, Russia
| | - Evgenii D Kryl'skii
- Department of Medical biochemistry and microbiology, Voronezh State University, Voronezh Russia -
| | - Konstantin K Shulgin
- Department of Medical biochemistry and microbiology, Voronezh State University, Voronezh Russia
| | - Ekaterina A Raskina
- Department of Hospital therapy and endocrinology, Voronezh State Medical University named after N.N. Burdenko, Voronezh, Russia
| | - Tatyana N Popova
- Department of Medical biochemistry and microbiology, Voronezh State University, Voronezh Russia
| | - Alexander N Pashkov
- Department of Biology, Voronezh State Medical University named after N.N. Burdenko, Voronezh, Russia
| | - Galina N Kuptsova
- Department of Hospital therapy and endocrinology, Voronezh State Medical University named after N.N. Burdenko, Voronezh, Russia
| |
Collapse
|
14
|
Zeng Z, Lan T, Wei Y, Wei X. CCL5/CCR5 axis in human diseases and related treatments. Genes Dis 2022; 9:12-27. [PMID: 34514075 PMCID: PMC8423937 DOI: 10.1016/j.gendis.2021.08.004] [Citation(s) in RCA: 110] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/08/2021] [Accepted: 08/12/2021] [Indexed: 02/05/2023] Open
Abstract
To defense harmful stimuli or maintain the immune homeostasis, the body produces and recruits a superfamily of cytokines such as interleukins, interferons, chemokines etc. Among them, chemokines act as crucial regulators in defense systems. CCL5/CCR5 combination is known for facilitating inflammatory responses, as well as inducing the adhesion and migration of different T cell subsets in immune responses. In addition, recent studies have shown that the interaction between CCL5 and CCR5 is involved in various pathological processes including inflammation, chronic diseases, cancers as well as the infection of COVID-19. This review focuses on how CCL5/CCR5 axis participates in the pathological processes of different diseases and their relevant signaling pathways for the regulation of the axis. Moreover, we highlighted the gene therapy and chemotherapy studies for treating CCR5-related diseases, including the ongoing clinical trials. The barriers and perspectives for future application and translational research were also summarized.
Collapse
Affiliation(s)
- Zhen Zeng
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, PR China
| | - Tianxia Lan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, PR China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, PR China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, PR China
| |
Collapse
|
15
|
Ding K, Li X, Ren X, Ding N, Tao L, Dong X, Chen Z. GBP5 promotes liver injury and inflammation by inducing hepatocyte apoptosis. FASEB J 2021; 36:e22119. [PMID: 34958688 DOI: 10.1096/fj.202101448r] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/02/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022]
Abstract
Liver injury is the first step in causing fibrosis, cirrhosis, and liver cancer, leading to mortality. However, the drivers of progressive liver injury are still incompletely defined. Here, we identify GBP5 as a major factor causing liver injury and inflammation. We show that the expression of GBP5 is abnormally elevated in the damaged liver, and its expression depends at least partially on the NF-κB-inducing kinase (NIK)/NF-κB2 signaling pathway. Knockout of Gbp5 ameliorates D-galactosamine/lipopolysaccharide (GalN/LPS)-induced liver injury and inflammation. Conversely, liver-specific overexpression of GBP5 induces liver injury and inflammation. Mechanistically, GBP5 induces hepatocyte apoptosis through the activation of both calpain/caspase 12/caspase 3 and TNFα/caspase 8/caspase 3 signaling pathways. Inhibition of either calpain activity or caspase 3 prevents GBP5-induced cell death. Our data demonstrate that GBP5 expression is induced by toxins or the NIK signaling pathway, which promotes both extrinsic and intrinsic apoptosis signaling pathways and further induces liver injury, providing a novel drug target for the treatment of liver injury and inflammation.
Collapse
Affiliation(s)
- Kaixin Ding
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xinzhi Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xiaomeng Ren
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, China.,Shenyang University of Chemical Technology, Shenyang, China
| | - Na Ding
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Li Tao
- 305 Hospital of People's Liberation Army, Beijing, China
| | - Xue Dong
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, China
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
16
|
Zhang L, Wang W, Liu L, Zhang Y, Zhang X. Peroxisome proliferator-activated receptor gamma coactivator 1-alpha protects a fibrotic liver from partial hepatectomy-induced advanced liver injury through regulating cell cycle arrest. Basic Clin Pharmacol Toxicol 2021; 130:254-267. [PMID: 34845850 PMCID: PMC9300180 DOI: 10.1111/bcpt.13697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/04/2021] [Accepted: 11/22/2021] [Indexed: 11/30/2022]
Abstract
Background A fibrotic liver may have an impaired regenerative capacity. Because liver transplantation is donor limited, understanding the regenerative ability of a fibrotic liver is important. Methods A two‐thirds partial hepatectomy (PH) was performed in C57Bl/6 mice with or without carbon tetrachloride (CCl4) treatment. Liver regeneration in the fibrotic liver after PH was assessed by the intrahepatic expression of the cell cycle regulators p53, p21, cyclin D1, c‐Fos and CDK2 using Western blot analysis. In addition, the expression of PGC‐1α and the cell proliferation‐related proteins PCNA and phosphate histone H3 was determined by Western blot and immunohistochemical staining analyses. Histone epigenetic modification of the PGC‐1α promoter was investigated through chromatin immunoprecipitation (ChIP) and reverse transcription–quantitative polymerase chain reaction (RT‐qPCR) assays. The impact of PGC‐1α on liver regeneration after PH was further evaluated in PGC‐1α‐knockout mice. Results A decreased expression of PGC‐1α and liver regeneration‐related genes in the fibrotic liver was detected after a PH. Histone acetylation at the PGC‐1α promoter led to increases in PGC‐1α expression and the survival rate in the fibrotic group after a PH. PGC‐1α‐mediated liver regeneration was further demonstrated in PGC‐1αf/falbcre+/0 mice. Conclusion Targeting PGC‐1α may represent a strategy to improve the treatment of PH in patients with liver fibrosis.
Collapse
Affiliation(s)
- Linzhong Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wei Wang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Lipeng Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yanghao Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiuying Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Liang R, Lin YH, Zhu H. Genetic and Cellular Contributions to Liver Regeneration. Cold Spring Harb Perspect Biol 2021; 14:a040832. [PMID: 34750173 PMCID: PMC9438780 DOI: 10.1101/cshperspect.a040832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The regenerative capabilities of the liver represent a paradigm for understanding tissue repair in solid organs. Regeneration after partial hepatectomy in rodent models is well understood, while regeneration in the context of clinically relevant chronic injuries is less studied. Given the growing incidence of fatty liver disease, cirrhosis, and liver cancer, interest in liver regeneration is increasing. Here, we will review the principles, genetics, and cell biology underlying liver regeneration, as well as new approaches being used to study heterogeneity in liver tissue maintenance and repair.
Collapse
Affiliation(s)
- Roger Liang
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Yu-Hsuan Lin
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Hao Zhu
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
18
|
Zhong X, Zhang Z, Shen H, Xiong Y, Shah YM, Liu Y, Fan X, Rui L. Hepatic NF-κB-Inducing Kinase and Inhibitor of NF-κB Kinase Subunit α Promote Liver Oxidative Stress, Ferroptosis, and Liver Injury. Hepatol Commun 2021; 5:1704-1720. [PMID: 34558831 PMCID: PMC8485893 DOI: 10.1002/hep4.1757] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/20/2021] [Accepted: 05/11/2021] [Indexed: 12/14/2022] Open
Abstract
Drug-induced hepatotoxicity limits development of new effective medications. Drugs and numerous endogenous/exogenous agents are metabolized/detoxified by hepatocytes, during which reactive oxygen species (ROS) are generated as a by-product. ROS has broad adverse effects on liver function and integrity, including damaging hepatocyte proteins, lipids, and DNA and promoting liver inflammation and fibrosis. ROS in concert with iron overload drives ferroptosis. Hepatic nuclear factor kappa B (NF-κB)-inducing kinase (NIK) is aberrantly activated in a broad spectrum of liver disease. NIK phosphorylates and activates inhibitor of NF-κB kinase subunit alpha (IKKα), and the hepatic NIK/IKKα cascade suppresses liver regeneration. However, the NIK/IKKα pathway has not been explored in drug-induced liver injury. Here, we identify hepatic NIK as a previously unrecognized mediator for acetaminophen (APAP)-induced acute liver failure. APAP treatment increased both NIK transcription and NIK protein stability in primary hepatocytes as well as in liver in mice. Hepatocyte-specific overexpression of NIK augmented APAP-induced liver oxidative stress in mice and increased hepatocyte death and mortality in a ROS-dependent manner. Conversely, hepatocyte-specific ablation of NIK or IKKα mitigated APAP-elicited hepatotoxicity and mortality. NIK increased lipid peroxidation and cell death in APAP-stimulated primary hepatocytes. Pretreatment with antioxidants or ferroptosis inhibitors blocked NIK/APAP-induced hepatocyte death. Conclusion: We unravel a previously unrecognized NIK/IKKα/ROS/ferroptosis axis engaged in liver disease progression.
Collapse
Affiliation(s)
- Xiao Zhong
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
- Department of Infectious DiseasesHunan Key Laboratory of Viral HepatitisXiangya HospitalCentral South UniversityChangshaChina
| | - Zhiguo Zhang
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Hong Shen
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Yi Xiong
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Yatrik M. Shah
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Yong Liu
- College of Life SciencesInstitute for Advanced StudiesWuhan UniversityWuhanChina
| | - Xue‐Gong Fan
- Department of Infectious DiseasesHunan Key Laboratory of Viral HepatitisXiangya HospitalCentral South UniversityChangshaChina
| | - Liangyou Rui
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
- Division of Gastroenterology and HepatologyDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
| |
Collapse
|
19
|
Role of NF-κB in Ageing and Age-Related Diseases: Lessons from Genetically Modified Mouse Models. Cells 2021; 10:cells10081906. [PMID: 34440675 PMCID: PMC8394846 DOI: 10.3390/cells10081906] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/17/2021] [Accepted: 07/23/2021] [Indexed: 12/21/2022] Open
Abstract
Ageing is a complex process, induced by multifaceted interaction of genetic, epigenetic, and environmental factors. It is manifested by a decline in the physiological functions of organisms and associated to the development of age-related chronic diseases and cancer development. It is considered that ageing follows a strictly-regulated program, in which some signaling pathways critically contribute to the establishment and maintenance of the aged state. Chronic inflammation is a major mechanism that promotes the biological ageing process and comorbidity, with the transcription factor NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) as a crucial mediator of inflammatory responses. This, together with the finding that the activation or inhibition of NF-κB can induce or reverse respectively the main features of aged organisms, has brought it under consideration as a key transcription factor that acts as a driver of ageing. In this review, we focused on the data obtained entirely through the generation of knockout and transgenic mouse models of either protein involved in the NF-κB signaling pathway that have provided relevant information about the intricate processes or molecular mechanisms that control ageing. We have reviewed the relationship of NF-κB and premature ageing; the development of cancer associated with ageing and the implication of NF-κB activation in the development of age-related diseases, some of which greatly increase the risk of developing cancer.
Collapse
|
20
|
Xiao W, He Z, Luo W, Feng D, Wang Y, Tang T, Yang A, Luo J. BYHWD Alleviates Inflammatory Response by NIK-Mediated Repression of the Noncanonical NF-κB Pathway During ICH Recovery. Front Pharmacol 2021; 12:632407. [PMID: 34025405 PMCID: PMC8138445 DOI: 10.3389/fphar.2021.632407] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/19/2021] [Indexed: 01/24/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is a life-threatening type of stroke that lacks effective treatments. The inflammatory response following ICH is a vital response that affects brain repair and organism recovery. The nuclear factor κB (NF-κB) signaling pathway is considered one of the most important inflammatory response pathways and one of its response pathways, the noncanonical NF-κB signaling pathway, is known to be associated with persistent effect and chronic inflammation. NF-κB–inducing kinase (NIK) via the noncanonical NF-κB signaling plays a key role in controlling inflammation. Here, we investigated potential effects of the traditional Chinese medicine formula Buyang Huanwu Decoction (BYHWD) on inflammatory response in a rat model of ICH recovery by inhibiting the NIK-mediated the noncanonical NF-κB signaling pathway. In the first part, rats were randomly divided into three groups: the sham group, the ICH group, and the BYHWD group. ICH was induced in rats by injecting collagenase (type VII) into the right globus pallidus of rats' brain. For the BYHWD group, rats were administered BYHWD (4.36 g/kg) once a day by intragastric administration until they were sacrificed. Neurological function was evaluated in rats by a modified neurological severity score (mNSS), the corner turn test, and the foot-fault test. The cerebral edema showed the degree of inflammatory response by sacrificed brain water content. Western blot and real-time quantitative reverse transcription PCR tested the activity of inflammatory response and noncanonical NF-κB signaling. In the second part, siRNA treatment and assessment of inflammation level as well as alterations in the noncanonical NF-κB signaling were performed to determine whether the effect of BYHWD on inflammatory response was mediated by suppression of NIK via the noncanonical NF-κB signaling pathway. We show that BYHWD treated rats exhibited: (i) better health conditions and better neural functional recovery; (ii) decreased inflammatory cytokine and the edema; (iii) reduced expression of NIK, a key protein in unregulated the noncanonical NF-κB signaling pathways; (iv) when compared with pretreated rats with NIK targeting (NIK siRNAs), showed the same effect of inhibiting the pathway and decreased inflammatory cytokine. BYHWD can attenuate the inflammatory response during ICH recovery in rats by inhibiting the NIK-mediated noncanonical NF-κB signaling pathway.
Collapse
Affiliation(s)
- Wei Xiao
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Zehui He
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, China
| | - Weikang Luo
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Dandan Feng
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Wang
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Tao Tang
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Ali Yang
- Department of Neurology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiekun Luo
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
21
|
Chen Q, Lu X, Zhang X. Noncanonical NF-κB Signaling Pathway in Liver Diseases. J Clin Transl Hepatol 2021; 9:81-89. [PMID: 33604258 PMCID: PMC7868705 DOI: 10.14218/jcth.2020.00063] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/04/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
The noncanonical NF-κB signaling pathway is an important branch of NF-κB signaling. It is involved in regulating multiple important biological processes, including inflammation and host immune response. A central adaptor protein of the noncanonical NF-κB pathway is NF-κB-inducing kinase (NIK), which activates the downstream kinase IKKα to process p100 to p52, thereby forming the RelB/p52 heterodimer to initiate the expression of target genes. Currently, many specific inhibitors and monoclonal antibodies targeting or triggering this pathway are being developed and tested for various diseases, including cancers, autoimmune diseases, and virus infection. Given that aberrant activation of the noncanonical NF-κB pathway is frequently observed in various liver diseases, targeting this pathway may be a promising therapeutic strategy to alleviate liver inflammation. Moreover, activation of this pathway may contribute to the antiviral immune response and promote the clearance of persistent hepatotropic virus infection. Here, we review the role of the noncanonical NF-κB pathway in the occurrence and development of different liver diseases, and discuss the potency and application of modulating the noncanonical NF-κB pathway for treatment of these liver diseases.
Collapse
Affiliation(s)
- Qianhui Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou,Guangdong, China
| | - Xinyu Lu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou,Guangdong, China
| | - Xiaoyong Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou,Guangdong, China
- Hepatology Unit, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
- Correspondence to: Xiaoyong Zhang, Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Guangzhou, Guangdong 510515, China. Tel: +86-20-62787830, E-mail:
| |
Collapse
|
22
|
Current etiological comprehension and therapeutic targets of acetaminophen-induced hepatotoxicity. Pharmacol Res 2020; 161:105102. [DOI: 10.1016/j.phrs.2020.105102] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/03/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023]
|
23
|
Zhao P, Saltiel AR. From overnutrition to liver injury: AMP-activated protein kinase in nonalcoholic fatty liver diseases. J Biol Chem 2020; 295:12279-12289. [PMID: 32651233 PMCID: PMC7443502 DOI: 10.1074/jbc.rev120.011356] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/24/2020] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver diseases (NAFLDs), especially nonalcoholic steatohepatitis (NASH), have become a major cause of liver transplant and liver-associated death. However, the pathogenesis of NASH is still unclear. Currently, there is no FDA-approved medication to treat this devastating disease. AMP-activated protein kinase (AMPK) senses energy status and regulates metabolic processes to maintain homeostasis. The activity of AMPK is regulated by the availability of nutrients, such as carbohydrates, lipids, and amino acids. AMPK activity is increased by nutrient deprivation and inhibited by overnutrition, inflammation, and hypersecretion of certain anabolic hormones, such as insulin, during obesity. The repression of hepatic AMPK activity permits the transition from simple steatosis to hepatocellular death; thus, activation might ameliorate multiple aspects of NASH. Here we review the pathogenesis of NAFLD and the impact of AMPK activity state on hepatic steatosis, inflammation, liver injury, and fibrosis during the transition of NAFL to NASH and liver failure.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Alan R Saltiel
- Department of Medicine, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA.
| |
Collapse
|
24
|
Li Z, Li X, Su MB, Gao LX, Zhou YB, Yuan B, Lyu X, Yan Z, Hu C, Zhang H, Luo C, Chen Z, Li J, Zhao Y. Discovery of a Potent and Selective NF-κB-Inducing Kinase (NIK) Inhibitor That Has Anti-inflammatory Effects in Vitro and in Vivo. J Med Chem 2020; 63:4388-4407. [PMID: 32216342 DOI: 10.1021/acs.jmedchem.0c00396] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The overexpression of NIK plays a critical role in liver inflammatory diseases. Treatment of such diseases with small-molecule NIK inhibitors is a reasonable but underexplored approach. In this paper, we reported the discovery of a potent and selective NIK inhibitor 46 (XT2). 46 inhibited the NIK kinase with an IC50 value of 9.1 nM in vitro, and it also potently suppressed NIK activities in intact cells. In isogenic primary hepatocytes, treatment of 46 efficiently suppressed the expressions of NIK-induced genes. 46 was orally bioavailable in mice with moderate systemic exposure. In a NIK-associated mouse liver inflammation model, 46 suppressed CCl4-induced upregulation of ALT, a key biomarker of acute liver injury. 46 also decreased immune cell infiltration into the injured liver tissue. Overall, these studies provide examples that an NIK inhibitor is able to suppress toxin-induced liver inflammations, which indicates its therapeutic potentials for the treatment of liver inflammatory diseases.
Collapse
Affiliation(s)
- Zhiqiang Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinzhi Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Ming-Bo Su
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Li-Xin Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Yu-Bo Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Bingchuan Yuan
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Xilin Lyu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Ziqin Yan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Chujiao Hu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
| | - Hao Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Cheng Luo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
- Open Studio for Druggability Research of Marine Natural Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), 1 Wenhai Road, Aoshanwei, Jimo, Qingdao 266237, China
| | - Yujun Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
25
|
Cigliola V, Ghila L, Chera S, Herrera PL. Tissue repair brakes: A common paradigm in the biology of regeneration. Stem Cells 2019; 38:330-339. [PMID: 31722129 DOI: 10.1002/stem.3118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 10/09/2019] [Accepted: 10/20/2019] [Indexed: 12/12/2022]
Abstract
To date, most attention on tissue regeneration has focused on the exploration of positive cues promoting or allowing the engagement of natural cellular restoration upon injury. In contrast, the signals fostering cell identity maintenance in the vertebrate body have been poorly investigated; yet they are crucial, for their counteraction could become a powerful method to induce and modulate regeneration. Here we review the mechanisms inhibiting pro-regenerative spontaneous adaptive cell responses in different model organisms and organs. The pharmacological or genetic/epigenetic modulation of such regenerative brakes could release a dormant but innate adaptive competence of certain cell types and therefore boost tissue regeneration in different situations.
Collapse
Affiliation(s)
- Valentina Cigliola
- Department of Cell Biology, Regeneration Next, Duke University Medical Center, Durham, North Carolina
| | - Luiza Ghila
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Simona Chera
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Pedro L Herrera
- Department of Genetic Medicine & Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
26
|
Valiño-Rivas L, Vaquero JJ, Sucunza D, Gutierrez S, Sanz AB, Fresno M, Ortiz A, Sanchez-Niño MD. NIK as a Druggable Mediator of Tissue Injury. Trends Mol Med 2019; 25:341-360. [PMID: 30926358 DOI: 10.1016/j.molmed.2019.02.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/07/2019] [Accepted: 02/13/2019] [Indexed: 12/15/2022]
Abstract
NF-κB-inducing kinase (NIK, MAP3K14) is best known as the apical kinase that triggers non-canonical NF-κB activation and by its role in the immune system. Recent data indicate a role for NIK expressed by non-lymphoid cells in cancer, kidney disease, liver injury, glucose homeostasis, osteosarcopenia, vascular calcification, hematopoiesis, and endothelial function. The spectrum of NIK-associated disease now ranges from immunodeficiency (when NIK is defective) to autoimmunity, cancer, sterile inflammation, fibrosis, and metabolic disease when NIK is overactive. The development of novel small-molecule NIK inhibitors has paved the way to test NIK targeting to treat disease in vivo, and may eventually lead to NIK targeting in the clinic. In addition, NIK activators are being explored for specific conditions such as myeloid leukemia.
Collapse
Affiliation(s)
- Lara Valiño-Rivas
- Department of Nephrology and Hypertension, Instituto de Investigación Sanitaria (IIS) Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid (UAM), Red de Investigación Renal (REDINREN), and Fundación Renal Íñigo Álvarez de Toledo (FRIAT), Madrid, Spain
| | - Juan José Vaquero
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcala and REDINREN, Madrid, Spain
| | - David Sucunza
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcala and REDINREN, Madrid, Spain
| | - Sara Gutierrez
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcala and REDINREN, Madrid, Spain
| | - Ana B Sanz
- Department of Nephrology and Hypertension, Instituto de Investigación Sanitaria (IIS) Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid (UAM), Red de Investigación Renal (REDINREN), and Fundación Renal Íñigo Álvarez de Toledo (FRIAT), Madrid, Spain
| | - Manuel Fresno
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas de la UAM, Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, Instituto de Investigación Sanitaria (IIS) Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid (UAM), Red de Investigación Renal (REDINREN), and Fundación Renal Íñigo Álvarez de Toledo (FRIAT), Madrid, Spain; These authors contributed equally.
| | - Maria Dolores Sanchez-Niño
- Department of Nephrology and Hypertension, Instituto de Investigación Sanitaria (IIS) Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid (UAM), Red de Investigación Renal (REDINREN), and Fundación Renal Íñigo Álvarez de Toledo (FRIAT), Madrid, Spain; These authors contributed equally.
| |
Collapse
|