1
|
Chen C, Bu X, Deng L, Xia J, Wang X, Chen L, Li W, Huang J, Chen Q, Wang C. Astragaloside IV as a promising therapeutic agent for liver diseases: current landscape and future perspectives. Front Pharmacol 2025; 16:1574154. [PMID: 40337517 PMCID: PMC12055773 DOI: 10.3389/fphar.2025.1574154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025] Open
Abstract
Astragaloside IV (C41H68O14, AS-IV) is a naturally occurring saponin isolated from the root of Astragalus membranaceus, a widely used traditional Chinese botanical drug in medicine. In recent years, AS-IV has attracted considerable attention for its hepatoprotective properties, which are attributed to its low toxicity as well as its anti-inflammatory, antioxidant and antitumour effects. Numerous preclinical studies have demonstrated its potential in the prevention and treatment of various liver diseases, including multifactorial liver injury, metabolic-associated fatty liver disease, liver fibrosis and liver cancer. Given the promising hepatoprotective potential of AS-IV and the growing interest in its research, this review provides a comprehensive summary of the current state of research on the hepatoprotective effects of AS-IV, based on literature available in databases such as CNKI, PubMed, ScienceDirect, Google Scholar and Web of Science. The hepatoprotective mechanisms of AS-IV are multifaceted, encompassing the inhibition of inflammatory responses, reduction of oxidative stress, improvement of insulin and leptin resistance, modulation of the gut microbiota, suppression of hepatocellular carcinoma cell proliferation and induction of tumour cell apoptosis. Notably, key molecular pathways involved in these effects include Nrf2/HO-1, NF-κB, NLRP3/Caspase-1, JNK/c-Jun/AP-1, PPARα/FSP1 and Akt/GSK-3β/β-catenin. Toxicity studies indicate that AS-IV has a high level of safety. In addition, this review discusses the sources, physicochemical properties, and current challenges in the development and clinical application of AS-IV, providing valuable insights into its potential as a hepatoprotective agent in the pharmaceutical and nutraceutical industries.
Collapse
Affiliation(s)
- Chunyan Chen
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xiaolan Bu
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Liping Deng
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jiayan Xia
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xinming Wang
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Li Chen
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wen Li
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jie Huang
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Qixiang Chen
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Cheng Wang
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
2
|
Meyer zu Schwabedissen A, Vergarajauregui S, Bertog M, Amann K, Engel FB, Daniel C. Protease-activated receptor 2 deficient mice develop less angiotensin II induced left ventricular hypertrophy but more cardiac fibrosis. PLoS One 2024; 19:e0310095. [PMID: 39637045 PMCID: PMC11620577 DOI: 10.1371/journal.pone.0310095] [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: 01/04/2024] [Accepted: 08/25/2024] [Indexed: 12/07/2024] Open
Abstract
AIMS Activation of Protease Activated Receptor 2 (PAR2) has been shown to be involved in regulation of injury-related processes including inflammation, fibrosis and hypertrophy. In this study we will investigate the role of PAR2 in cardiac injury in a mouse model of hypertension using continuous infusion with angiotensin II. METHODS Hypertension was induced in 12 weeks old wildtype (wt, n = 8) and PAR2 deficient mice (n = 9) by continuous infusion with angiotensin II for 4 weeks using osmotic minipumps. At the end, hearts were collected for analysis of left ventricular hypertrophy (LVH), myocardial capillary supply, fibrosis and localization of PAR2 expression using histological, immunohistological and mRNA expression analysis techniques. In addition, rat cardiac fibroblasts were treated with angiotensin II and PAR2 was inhibited by a blocking antibody and the PAR2 inhibitor AZ3451. RESULTS Cardiac PAR2 mRNA expression was downregulated by 40±20% in wt mice treated with AngII compared to untreated controls. Four weeks after AngII treatment, LVH was significantly increased in AngII-treated wt mice compared to similarly treated PAR2-deficient animals as determined by relative heart weight, left ventricular cross-sectional area, and analysis of ventricular lumen area determined on sections. Treatment of wt mice resulted in an approximately 3-fold increase in cardiac expression of FGF23, which was 50% lower in PAR2-deficient animals compared to wt animals and therefore no longer significantly different from expression levels in untreated control mice. In contrast, cardiac interstitial fibrosis was significantly higher in PAR2-deficient mice compared to similar treated wt controls, as assessed by Sirius Red staining (>3-fold) and collagen IV staining (>2-fold). Additional experiments with isolated cardiac fibroblasts showed induction of pro-fibrotic genes when treated with PAR2 inhibitors. CONCLUSION In angiotensin II-induced cardiac injury, PAR2 deficiency has an ambivalent effect, enhancing fibrosis on the one hand, but reducing LVH on the other.
Collapse
Affiliation(s)
- Albrecht Meyer zu Schwabedissen
- Department of Nephropathology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Silvia Vergarajauregui
- Department of Nephropathology, Institute of Pathology and Department of Cardiology, Experimental Renal and Cardiovascular Research, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Marko Bertog
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Kerstin Amann
- Department of Nephropathology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Felix B. Engel
- Department of Nephropathology, Institute of Pathology and Department of Cardiology, Experimental Renal and Cardiovascular Research, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christoph Daniel
- Department of Nephropathology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
3
|
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
|
4
|
Fan M, Fan X, Lai Y, Chen J, Peng Y, Peng Y, Xiang L, Ma Y. Protease-Activated Receptor 2 in inflammatory skin disease: current evidence and future perspectives. Front Immunol 2024; 15:1448952. [PMID: 39301020 PMCID: PMC11410643 DOI: 10.3389/fimmu.2024.1448952] [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: 06/14/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024] Open
Abstract
Protease-activated receptor-2 (PAR2) is a class-A G protein-coupled receptor (GPCR) activated by serine proteases and is expressed by multiple tissues, including the skin. PAR2 is involved in the skin inflammatory response, promoting Th2 inflammation, delaying skin barrier repair, and affecting the differentiation of keratinocytes. It also participates in the transmission of itch and pain sensations in the skin. Increasing evidence indicates that PAR2 plays an important role in the pathogenesis of inflammatory skin diseases such as acne vulgaris, rosacea, psoriasis, and atopic dermatitis. Additional focus will be placed on potential targeted therapies based on PAR2. The Goal of this review is to outline the emerging effects of PAR2 activation in inflammatory skin disease and highlight the promise of PAR2 modulators.
Collapse
Affiliation(s)
- Mengjie Fan
- Department of Dermatology, Huashan Hosptial, Fudan University, Shanghai, China
| | - Xiaoyao Fan
- Department of Dermatology, Huashan Hosptial, Fudan University, Shanghai, China
| | - Yangfan Lai
- Department of Dermatology, Huashan Hosptial, Fudan University, Shanghai, China
| | - Jin Chen
- Department of Dermatology, Huashan Hosptial, Fudan University, Shanghai, China
| | - Yifan Peng
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Yao Peng
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Leihong Xiang
- Department of Dermatology, Huashan Hosptial, Fudan University, Shanghai, China
| | - Ying Ma
- Department of Dermatology, Huashan Hosptial, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Oshins R, Greenberg Z, Tai YL, Zhao D, Wang X, Mehrad B, He M, Patel I, Khartabil L, Zhou H, Brantly M, Khodayari N. Extracellular Vesicle-Associated Neutrophil Elastase Activates Hepatic Stellate Cells and Promotes Liver Fibrogenesis via ERK1/2 Pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.608832. [PMID: 39229038 PMCID: PMC11370372 DOI: 10.1101/2024.08.20.608832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Liver fibrosis associated with increased mortality is caused by activation of hepatic stellate cells and excessive production and accumulation of extracellular matrix in response to fibrotic insults. It has been shown that in addition to liver inflammation, systemic inflammation also contributes to liver fibrogenesis. A deeper understanding of mechanisms that control liver fibrotic response to intra- and extra-hepatic inflammation is essential to develop novel clinical strategies against this disease. Extracellular vesicles (EV) have been recognized as immune mediators that facilitate activation of hepatic stellate cells. In inflammatory diseases, activated neutrophils release neutrophil elastase (NE) bound to EV, which has been identified as a significant contributor to inflammation by promoting immune cell activation. Here, we aimed to explore the role of inflammation derived plasma EV-associated NE in liver fibrogenesis and its potential mechanisms. We show EV-associated NE induces activation, proliferation and migration of hepatic stellate cells by promoting activation of the ERK1/2 signaling pathway. This effect did not occur through EV without surface NE, and Sivelestat, a NE inhibitor, inhibited activation of the ERK1/2 signaling pathway mediated by EV-associated NE. Moreover, we found plasma EV-associated NE increases deposition of collagen1 and α-smooth muscle actin in the liver of a mouse model of liver fibrosis (Mdr2-/-). Notably, this effect does not occur in control mice without preexisting liver disease. These data suggest that EV-associated NE is a pro-fibrogenic factor for hepatic stellate cell activation via the ERK1/2 signaling pathway in pre-existing liver injuries. Inhibition of the plasma EV-associated NE in inflammatory conditions may be a therapeutic target for liver fibrosis in patients with inflammatory diseases.
Collapse
Affiliation(s)
- Regina Oshins
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine; University of Florida, Gainesville, Florida, USA
| | - Zachary Greenberg
- Department of Pharmaceutics, College of Pharmacy; University of Florida, Gainesville, Florida, USA
| | - Yun-Ling Tai
- Department of Microbiology and Immunology; Virginia Commonwealth University, Richmond VA Medical Center, Richmond, Virginia, USA
| | - Derrick Zhao
- Department of Microbiology and Immunology; Virginia Commonwealth University, Richmond VA Medical Center, Richmond, Virginia, USA
| | - Xuan Wang
- Department of Microbiology and Immunology; Virginia Commonwealth University, Richmond VA Medical Center, Richmond, Virginia, USA
| | - Borna Mehrad
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine; University of Florida, Gainesville, Florida, USA
| | - Mei He
- Department of Pharmaceutics, College of Pharmacy; University of Florida, Gainesville, Florida, USA
| | - Ishan Patel
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine; University of Florida, Gainesville, Florida, USA
| | - Laith Khartabil
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine; University of Florida, Gainesville, Florida, USA
| | - Huiping Zhou
- Department of Microbiology and Immunology; Virginia Commonwealth University, Richmond VA Medical Center, Richmond, Virginia, USA
| | - Mark Brantly
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine; University of Florida, Gainesville, Florida, USA
| | - Nazli Khodayari
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine; University of Florida, Gainesville, Florida, USA
| |
Collapse
|
6
|
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
|
7
|
Huang Y, Chen L, Li L, Qi Y, Tong H, Wu H, Xu J, Leng L, Cheema S, Sun G, Xia Z, McGuire J, Rodrigues B, Young LH, Bucala R, Qi D. Downregulation of adipose LPL by PAR2 contributes to the development of hypertriglyceridemia. JCI Insight 2024; 9:e173240. [PMID: 38973609 PMCID: PMC11383372 DOI: 10.1172/jci.insight.173240] [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: 06/21/2023] [Accepted: 05/22/2024] [Indexed: 07/09/2024] Open
Abstract
Lipoprotein lipase (LPL) hydrolyzes circulating triglycerides (TGs), releasing fatty acids (FA) and promoting lipid storage in white adipose tissue (WAT). However, the mechanisms regulating adipose LPL and its relationship with the development of hypertriglyceridemia are largely unknown. WAT from obese humans exhibited high PAR2 expression, which was inversely correlated with the LPL gene. Decreased LPL expression was also inversely correlated with elevated plasma TG levels, suggesting that adipose PAR2 might regulate hypertriglyceridemia by downregulating LPL. In mice, aging and high palmitic acid diet (PD) increased PAR2 expression in WAT, which was associated with a high level of macrophage migration inhibitory factor (MIF). MIF downregulated LPL expression and activity in adipocytes by binding with CXCR2/4 receptors and inhibiting Akt phosphorylation. In a MIF overexpression model, high-circulating MIF levels suppressed adipose LPL, and this suppression was associated with increased plasma TGs but not FA. Following PD feeding, adipose LPL expression and activity were significantly reduced, and this reduction was reversed in Par2-/- mice. Recombinant MIF infusion restored high plasma MIF levels in Par2-/- mice, and the levels decreased LPL and attenuated adipocyte lipid storage, leading to hypertriglyceridemia. These data collectively suggest that downregulation of adipose LPL by PAR2/MIF may contribute to the development of hypertriglyceridemia.
Collapse
Affiliation(s)
- Yiheng Huang
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Liujun Chen
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lisha Li
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yadan Qi
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Haibin Tong
- College of Life and Environment Sciences, Wenzhou University, Wenzhou, Zhejiang, China
| | - Hong Wu
- Institute of Cardiovascular Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jinjie Xu
- Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Lin Leng
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Guang Sun
- Faculty of Medicine, Memorial University, St. John’s, Newfoundland, Canada
| | - Zhengyuan Xia
- Guangdong Medical University, Zhanjiang, Guangdong, China
| | - John McGuire
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Brian Rodrigues
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lawrence H. Young
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dake Qi
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Faculty of Medicine, Memorial University, St. John’s, Newfoundland, Canada
| |
Collapse
|
8
|
Adams F, Zimmermann CM, Baldassi D, Pehl TM. Pulmonary siRNA Delivery with Sophisticated Amphiphilic Poly(Spermine Acrylamides) for the Treatment of Lung Fibrosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308775. [PMID: 38126895 PMCID: PMC7616748 DOI: 10.1002/smll.202308775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/16/2023] [Indexed: 12/23/2023]
Abstract
RNA interference (RNAi) is an efficient strategy to post-transcriptionally silence gene expression. While all siRNA drugs on the market target the liver, the lung offers a variety of currently undruggable targets, which can potentially be treated with RNA therapeutics. To achieve this goal, the synthesis of poly(spermine acrylamides) (P(SpAA) is reported herein. Polymers are prepared via polymerization of N-acryloxysuccinimide (NAS) and afterward this active ester is converted into spermine-based pendant groups. Copolymerizations with decylacrylamide are employed to increase the hydrophobicity of the polymers. After deprotection, polymers show excellent siRNA encapsulation to obtain perfectly sized polyplexes at very low polymer/RNA ratios. In vitro 2D and 3D cell culture, ex vivo and in vivo experiments reveal superior properties of amphiphilic spermine-copolymers with respect to delivery of siRNA to lung cells in comparison to commonly used lipid-based transfection agents. In line with the in vitro results, siRNA delivery to human lung explants confirm more efficient gene silencing of protease-activated receptor 2 (PAR2), a G protein-coupled receptor involved in fibrosis. This study reveals the importance of the balance between efficient polyplex formation, cellular uptake, gene knockdown, and toxicity for efficient siRNA delivery in vitro, in vivo, and in fibrotic human lung tissue ex vivo.
Collapse
Affiliation(s)
- Friederike Adams
- Pharmaceutical Technology and Biopharmaceutics, Department Pharmacy Ludwig-Maximilians-University Munich, Butenandtstr. 5−13, 81377Munich, Germany
- Institute of Polymer Chemistry Chair of Macromolecular Materials and Fiber Chemistry, University of Stuttgart, Pfaffenwaldring 55, 70569Stuttgart, Germany
- Center for Ophthalmology University Eye Hospital Tübingen, Elfriede-Aulhorn-Straße 7, 72076 Tübingen, Germany
| | | | - Domizia Baldassi
- Pharmaceutical Technology and Biopharmaceutics, Department Pharmacy Ludwig-Maximilians-University Munich, Butenandtstr. 5−13, 81377Munich, Germany
| | - Thomas M. Pehl
- WACKER-Chair of Macromolecular Chemistry, Catalysis Research Center, Department of Chemistry, Technical University Munich, Lichtenbergstr. 4, 85748Garching bei München, Germany
| |
Collapse
|
9
|
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
|
10
|
Lee HY, You DJ, Taylor-Just A, Tisch LJ, Bartone RD, Atkins HM, Ralph LM, Antoniak S, Bonner JC. Role of the protease-activated receptor-2 (PAR2) in the exacerbation of house dust mite-induced murine allergic lung disease by multi-walled carbon nanotubes. Part Fibre Toxicol 2023; 20:32. [PMID: 37580758 PMCID: PMC10424461 DOI: 10.1186/s12989-023-00538-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 06/28/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Pulmonary exposure to multi-walled carbon nanotubes (MWCNTs) has been reported to exert strong pro-inflammatory and pro-fibrotic adjuvant effects in mouse models of allergic lung disease. However, the molecular mechanisms through which MWCNTs exacerbate allergen-induced lung disease remain to be elucidated. We hypothesized that protease-activated receptor 2 (PAR2), a G-protein coupled receptor previously implicated in the pathogenesis of various diseases including pulmonary fibrosis and asthma, may play an important role in the exacerbation of house dust mite (HDM) allergen-induced lung disease by MWCNTs. METHODS Wildtype (WT) male C57BL6 mice and Par2 KO mice were exposed to vehicle, MWCNTs, HDM extract, or both via oropharyngeal aspiration 6 times over a period of 3 weeks and were sacrificed 3-days after the final exposure (day 22). Bronchoalveolar lavage fluid (BALF) was harvested to measure changes in inflammatory cells, total protein, and lactate dehydrogenase (LDH). Lung protein and RNA were assayed for pro-inflammatory or profibrotic mediators, and formalin-fixed lung sections were evaluated for histopathology. RESULTS In both WT and Par2 KO mice, co-exposure to MWCNTs synergistically increased lung inflammation assessed by histopathology, and increased BALF cellularity, primarily eosinophils, as well as BALF total protein and LDH in the presence of relatively low doses of HDM extract that alone produced little, if any, lung inflammation. In addition, both WT and par2 KO mice displayed a similar increase in lung Cc1-11 mRNA, which encodes the eosinophil chemokine CCL-11, after co-exposure to MWCNTs and HDM extract. However, Par2 KO mice displayed significantly less airway fibrosis as determined by quantitative morphometry compared to WT mice after co-exposure to MWCNTs and HDM extract. Accordingly, at both protein and mRNA levels, the pro-fibrotic mediator arginase 1 (ARG-1), was downregulated in Par2 KO mice exposed to MWCNTs and HDM. In contrast, phosphorylation of the pro-inflammatory transcription factor NF-κB and the pro-inflammatory cytokine CXCL-1 was increased in Par2 KO mice exposed to MWCNTs and HDM. CONCLUSIONS Our study indicates that PAR2 mediates airway fibrosis but not eosinophilic lung inflammation induced by co-exposure to MWCNTs and HDM allergens.
Collapse
Affiliation(s)
- Ho Young Lee
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Dorothy J You
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Alexia Taylor-Just
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Logan J Tisch
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Ryan D Bartone
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Hannah M Atkins
- Department of Population Health and Pathobiology, North Carolina State University, Raleigh, NC, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Lauren M Ralph
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Silvio Antoniak
- UNC Blood Research Center, Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - James C Bonner
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
11
|
Yang X, Ou Y, Yang Y, Wang L, Zhang Y, Zhao F, Shui P, Qing J. Targeting endothelial coagulation signaling ameliorates liver obstructive cholestasis and dysfunctional angiogenesis. Exp Biol Med (Maywood) 2023; 248:1242-1253. [PMID: 37644866 PMCID: PMC10621472 DOI: 10.1177/15353702231191190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/08/2023] [Indexed: 08/31/2023] Open
Abstract
Cholestatic fibrogenesis is a pathobiological process in which cumulative injury to the bile ducts coincides with progressive liver fibrosis. The pathobiologic mechanisms underlying fibrogenesis and disease progression remain poorly understood. Currently, there is no effective treatment for liver fibrosis. In this study, we discovered that components of the coagulation cascade were associated with the advanced progression of obstructive cholestasis, and anticoagulant therapy could improve liver cholestasis-induced fibrosis. In a mouse model of common bile duct ligation (BDL), which mimics cholestatic liver injury, RNA sequencing analysis revealed an increased expression of coagulation factors in endothelial cells. Pharmacological targeting of the coagulation signaling by hirudin, an anticoagulatory antagonist of thrombin, ameliorated obstructive cholestasis, and attenuated liver fibrosis symptoms. Hirudin attenuated fibrosis-associated angiogenesis, endothelial-to-mesenchymal transition (EndMT), and tissue hypoxia and reduced liver inflammation after BDL. Furthermore, hirudin suppressed YAP (Yes-associated protein) signaling and its downstream effectors in vascular endothelial cells, which are considered with profibrotic characteristics. In conclusion, we demonstrated that pharmacological targeting of coagulation signaling by hirudin has the potential to alleviate liver obstructive cholestasis and fibrosis.
Collapse
Affiliation(s)
- Xue Yang
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
- Department of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Yangying Ou
- National Traditional Chinese Medicine Clinical Research Base and Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Ying Yang
- National Traditional Chinese Medicine Clinical Research Base and Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Lu Wang
- National Traditional Chinese Medicine Clinical Research Base and Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yuwei Zhang
- National Traditional Chinese Medicine Clinical Research Base and Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Fulan Zhao
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Pixian Shui
- Department of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Jie Qing
- National Traditional Chinese Medicine Clinical Research Base and Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| |
Collapse
|
12
|
Costa CRC, Belchor MN, Roggero A, Moraes LL, Samelo R, Annunciato I, Bonturi CR, Oliva MLV, Sousa SF, de Oliveira MA, Toyama MH. The First Anti-Snakebite and Hepatoprotective Characterization of a Trypsin Kunitz-like Inhibitor (EcTI) from the Plant Enterolobium contortisiliquum; A Case of Two Soul Mates Meeting. Pharmaceuticals (Basel) 2023; 16:ph16040632. [PMID: 37111388 PMCID: PMC10145096 DOI: 10.3390/ph16040632] [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: 02/28/2023] [Revised: 04/06/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Snake venom serine protease (SVSP) interferes with the regulation and control of important biological reactions in homeostasis and can be classified as an activator of the fibrinolytic system and platelet aggregation. Our group has recently isolated a new serine protease from Crotalus durissus terrificus total venom (Cdtsp-2). This protein exhibits edematogenic capacity and myotoxic activity. A Kunitz-like EcTI inhibitor protein with a molecular mass of 20 kDa was isolated from Enterolobium contortisiliquum and showed high trypsin inhibition. Thus, the objective of this work is to verify the possible inhibition of the pharmacological activities of Cdtsp-2 by the Kutinz-type inhibitor EcTI. To isolate Cdtsp-2 from total C. d. terrificus venom, we used three-step chromatographic HPLC. Using the mice paw edema model, we observed an edematogenic effect, myotoxicity and hepatotoxicity caused by Cdtsp-2. In vitro and in vivo experiments showed that the alterations in hemostasis caused by Cdtsp-2 are crucial for the development of marked hepatotoxicity and that EcTI significantly inhibits the enzymatic and pharmacological activities of Cdtsp-2. Kunitz-like inhibitor may be a viable alternative for the development of ancillary treatments against the biological activities of venoms.
Collapse
Affiliation(s)
- Caroline R C Costa
- Center of Natural and Human Sciences, Federal University of ABC (UFABC), Santo André 09210-580, SP, Brazil
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| | - Mariana N Belchor
- Center of Natural and Human Sciences, Federal University of ABC (UFABC), Santo André 09210-580, SP, Brazil
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| | - Airam Roggero
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| | - Laila L Moraes
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| | - Ricardo Samelo
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| | - Isabelly Annunciato
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| | - Camila R Bonturi
- National Institute of Pharmacology (INFAR), Federal University of São Paulo (UNIFESP), São Paulo 04044-020, SP, Brazil
| | - Maria L V Oliva
- National Institute of Pharmacology (INFAR), Federal University of São Paulo (UNIFESP), São Paulo 04044-020, SP, Brazil
| | - Sergio F Sousa
- Unit of Applied Biomolecular Sciences (UCIBIO), REQUIMTE-BioSIM-Medicine Faculty, Porto University, 4050-345 Porto, Portugal
| | - Marcos A de Oliveira
- Center of Natural and Human Sciences, Federal University of ABC (UFABC), Santo André 09210-580, SP, Brazil
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| | - Marcos H Toyama
- Center of Natural and Human Sciences, Federal University of ABC (UFABC), Santo André 09210-580, SP, Brazil
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| |
Collapse
|
13
|
Shpakov AO. Allosteric Regulation of G-Protein-Coupled Receptors: From Diversity of Molecular Mechanisms to Multiple Allosteric Sites and Their Ligands. Int J Mol Sci 2023; 24:6187. [PMID: 37047169 PMCID: PMC10094638 DOI: 10.3390/ijms24076187] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Allosteric regulation is critical for the functioning of G protein-coupled receptors (GPCRs) and their signaling pathways. Endogenous allosteric regulators of GPCRs are simple ions, various biomolecules, and protein components of GPCR signaling (G proteins and β-arrestins). The stability and functional activity of GPCR complexes is also due to multicenter allosteric interactions between protomers. The complexity of allosteric effects caused by numerous regulators differing in structure, availability, and mechanisms of action predetermines the multiplicity and different topology of allosteric sites in GPCRs. These sites can be localized in extracellular loops; inside the transmembrane tunnel and in its upper and lower vestibules; in cytoplasmic loops; and on the outer, membrane-contacting surface of the transmembrane domain. They are involved in the regulation of basal and orthosteric agonist-stimulated receptor activity, biased agonism, GPCR-complex formation, and endocytosis. They are targets for a large number of synthetic allosteric regulators and modulators, including those constructed using molecular docking. The review is devoted to the principles and mechanisms of GPCRs allosteric regulation, the multiplicity of allosteric sites and their topology, and the endogenous and synthetic allosteric regulators, including autoantibodies and pepducins. The allosteric regulation of chemokine receptors, proteinase-activated receptors, thyroid-stimulating and luteinizing hormone receptors, and beta-adrenergic receptors are described in more detail.
Collapse
Affiliation(s)
- Alexander O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| |
Collapse
|
14
|
Wang GY, Zhang XY, Wang CJ, Guan YF. Emerging novel targets for nonalcoholic fatty liver disease treatment: Evidence from recent basic studies. World J Gastroenterol 2023; 29:75-95. [PMID: 36683713 PMCID: PMC9850950 DOI: 10.3748/wjg.v29.i1.75] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/29/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), a leading chronic disease worldwide, affects approximately a quarter of the global population. Nonalcoholic steatohepatitis (NASH) is an advanced form of NAFLD and is more likely to progress to liver fibrosis than simple steatosis. NASH is also identified as the most rapidly growing cause of hepatocellular carcinoma. Although in the past decade, several phase II/III clinical trials have shown promising results in the use of novel drugs targeting lipid synthase, farnesoid X receptor signaling, peroxisome proliferator-activated receptor signaling, hepatocellular injury, and inflammatory signaling, proven pharmaceutical agents to treat NASH are still lacking. Thus, continuous exploration of the mechanism underlying the pathogenesis of NAFLD and the identification of novel therapeutic targets remain urgent tasks in the field. In the current review, we summarize studies reported in recent years that not only provide new insights into the mechanisms of NAFLD development but also explore the possibility of treating NAFLD by targeting newly identified signaling pathways. We also discuss evidence focusing on the intrahepatic targets involved in the pathogenesis of NAFLD as well as extrahepatic targets affecting liver metabolism and function.
Collapse
Affiliation(s)
- Guang-Yan Wang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin 300070, China
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin 300070, China
| | - Xiao-Yan Zhang
- Health Science Center, East China Normal University, Shanghai 200241, China
| | - Chun-Jiong Wang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin 300070, China
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin 300070, China
| | - You-Fei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning Province, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning Province, China
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian 116044, Liaoning Province, China
| |
Collapse
|
15
|
Ye X, Li J, Liu Z, Sun X, Wei D, Song L, Wu C. Peptide mediated therapy in fibrosis: Mechanisms, advances and prospects. Biomed Pharmacother 2023; 157:113978. [PMID: 36423541 DOI: 10.1016/j.biopha.2022.113978] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/30/2022] [Accepted: 11/03/2022] [Indexed: 11/22/2022] Open
Abstract
Fibrosis, a disease characterized by an excess accumulation of extracellular matrix components, could lead to organ failure and death, and is to blame for up to 45 % of all fatalities in developed nations. These disorders all share the common trait of an unchecked and increasing accumulation of fibrotic tissue in the affected organs, which leads to their malfunction and eventual failure, even if their underlying causes are highly diverse and, in some cases, remain unclear. Numerous studies have identified activated myofibroblasts as the common cellular elements ultimately responsible for the replacement of normal tissues with nonfunctional fibrotic tissue. The transforming growth factor-β pathway, for instance, plays a significant role in practically all kinds of fibrosis. However, there is no specific drug for the treatment of fibrosis, several medications with anti-hepatic fibrosis properties are still in the research and development stages. Peptide, which refers to a substance consisting of 2-50 amino acids, is characterized by structural diversity, low toxicity, biological activities, easy absorption, specific targeting, few side effects, and has been proven to be effective in anti-fibrosis. Here, we summarized various anti-fibrosis peptides in fibrosis including the liver, lungs, kidneys, and other organs. This review will provide a new insight into peptide mediated anti-fibrosis and is helpful to creation of antifibrotic medications.
Collapse
Affiliation(s)
- Xun Ye
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Jinhu Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Zibo Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Xue Sun
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Daneng Wei
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, PR China.
| | - Chunjie Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| |
Collapse
|
16
|
Shearer AM, Wang Y, Fletcher EK, Rana R, Michael ES, Nguyen N, Abdelmalek MF, Covic L, Kuliopulos A. PAR2 promotes impaired glucose uptake and insulin resistance in NAFLD through GLUT2 and Akt interference. Hepatology 2022; 76:1778-1793. [PMID: 35603482 PMCID: PMC9669194 DOI: 10.1002/hep.32589] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/28/2022] [Accepted: 05/13/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Insulin resistance and poor glycemic control are key drivers of the development of NAFLD and have recently been shown to be associated with fibrosis progression in NASH. However, the underlying mechanisms involving dysfunctional glucose metabolism and relationship with NAFLD/NASH progression remain poorly understood. We set out to determine whether protease-activated receptor 2 (PAR2), a sensor of extracellular inflammatory and coagulation proteases, links NAFLD and NASH with liver glucose metabolism. APPROACH AND RESULTS Here, we demonstrate that hepatic expression of PAR2 increases in patients and mice with diabetes and NAFLD/NASH. Mechanistic studies using whole-body and liver-specific PAR2-knockout mice reveal that hepatic PAR2 plays an unexpected role in suppressing glucose internalization, glycogen storage, and insulin signaling through a bifurcating Gq -dependent mechanism. PAR2 activation downregulates the major glucose transporter of liver, GLUT2, through Gq -MAPK-FoxA3 and inhibits insulin-Akt signaling through Gq -calcium-CaMKK2 pathways. Therapeutic dosing with a liver-homing pepducin, PZ-235, blocked PAR2-Gq signaling and afforded significant improvements in glycemic indices and HbA1c levels in severely diabetic mice. CONCLUSIONS This work provides evidence that PAR2 is a major regulator of liver glucose homeostasis and a potential target for the treatment of diabetes and NASH.
Collapse
Affiliation(s)
- Andrew M. Shearer
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Boston, Massachusetts, USA
- Tufts University School of Graduate Biomedical Sciences/DMCB, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Yanling Wang
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Boston, Massachusetts, USA
| | - Elizabeth K. Fletcher
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Boston, Massachusetts, USA
| | - Rajashree Rana
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Boston, Massachusetts, USA
| | - Emily S. Michael
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Boston, Massachusetts, USA
| | - Nga Nguyen
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Boston, Massachusetts, USA
| | - Manal F. Abdelmalek
- Division of Gastroenterology and Hepatology, Duke University Medical Center, Durham, North Carolina, USA
| | - Lidija Covic
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Boston, Massachusetts, USA
- Tufts University School of Graduate Biomedical Sciences/DMCB, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Athan Kuliopulos
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Boston, Massachusetts, USA
- Tufts University School of Graduate Biomedical Sciences/DMCB, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Zhang H, Ren L, Shivnaraine RV. Targeting GPCRs to treat cardiac fibrosis. Front Cardiovasc Med 2022; 9:1011176. [PMID: 36277752 PMCID: PMC9582444 DOI: 10.3389/fcvm.2022.1011176] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiac fibrosis occurs ubiquitously in ischemic heart failure, genetic cardiomyopathies, diabetes mellitus, and aging. It triggers myocardial stiffness, which impairs cardiac function, ultimately progressing to end-stage heart failure and increased mortality. Although several targets for anti-fibrotic therapies have been identified, including TGF-β and receptor tyrosine kinase, there is currently no FDA-approved drug specifically targeting cardiac fibrosis. G protein-coupled receptors (GPCRs) are integral, multipass membrane-bound receptors that exhibit diverse and cell-specific expression, offering novel and unrealized therapeutic targets for cardiac fibrosis. This review highlights the emerging roles of several GPCRs and briefly explores their downstream pathways that are crucial in cardiac fibrosis. We will not only provide an overview of the GPCRs expressed on cardiac fibroblasts that are directly involved in myofibroblast activation but also describe those GPCRs which contribute to cardiac fibrosis via indirect crosstalk mechanisms. We also discuss the challenges of identifying novel effective therapies for cardiac fibrosis and offer strategies to circumvent these challenges.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Medicine, Division of Cardiovascular Medicine, Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States,*Correspondence: Hao Zhang
| | - Lu Ren
- Department of Medicine, Division of Cardiovascular Medicine, Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | | |
Collapse
|
18
|
Xu H, Tilley DG. Pepducin-mediated G Protein-Coupled Receptor Signaling in the Cardiovascular System. J Cardiovasc Pharmacol 2022; 80:378-385. [PMID: 35170495 PMCID: PMC9365886 DOI: 10.1097/fjc.0000000000001236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/29/2022] [Indexed: 01/31/2023]
Abstract
ABSTRACT Pepducins are small-lipidated peptides designed from the intracellular loops of G protein-coupled receptors (GPCRs) that act in an allosteric manner to modulate the activity of GPCRs. Over the past 2 decades, pepducins have progressed initially from pharmacologic tools used to manipulate GPCR activity in an orthosteric site-independent manner to compounds with therapeutic potential that have even been used safely in phase 1 and 2 clinical trials in human subjects. The effect of pepducins at their cognate receptors has been shown to vary between antagonist, partial agonist, and biased agonist outcomes in various primary and clonal cell systems, with even small changes in amino acid sequence altering these properties and their receptor selectivity. To date, pepducins designed from numerous GPCRs have been studied for their impact on pathologic conditions, including cardiovascular diseases such as thrombosis, myocardial infarction, and atherosclerosis. This review will focus in particular on pepducins designed from protease-activated receptors, C-X-C motif chemokine receptors, formyl peptide receptors, and the β2-adrenergic receptor. We will discuss the historic context of pepducin development for each receptor, as well as the structural, signaling, pathophysiologic consequences, and therapeutic potential for each pepducin class.
Collapse
Affiliation(s)
- Heli Xu
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | | |
Collapse
|
19
|
Blockade of protease-activated receptor 2 (PAR-2) attenuates vascular dyshomeostasis and liver dysfunction induced by dengue virus infection. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
20
|
Li B, Li Y, Li S, Li H, Liu L, Xu Y. Inhibition of Protease Activated Receptor 2 Attenuates HBx-Induced Inflammation and Mitochondria Oxidative Stress. Infect Drug Resist 2022; 15:961-973. [PMID: 35299854 PMCID: PMC8921841 DOI: 10.2147/idr.s343864] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 02/19/2022] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) infection is one of the global public problems. Among the known infection cases, HBV X protein (HBx) is one of the key inducements of viral replication and host infection. This study was aimed to uncover the role of protease activated receptor 2 (PAR2) on HBx-induced liver injury. METHODS A PAR2-KO mouse model expressing HBx was constructed using hydrodynamics-based in vivo gene transfection method. In addition, pcDNA3.1-HBx was used to over-express HBx in LO2 cells. The effects of HBx overexpression on inflammation and mitochondria oxidative stress were evaluated. RESULTS We found that PAR2 protein level was increased by HBx overexpression. The enforced HBx inhibited LO2 cells apoptosis. Meanwhile, HBx induced inflammation reactions through promoting the secretion of pro-inflammatory cytokines such as TNF-α, IL-6, and CXCL-2. Overexpressed HBx also resulted in mitochondria oxidative stress by upregulation of ROS level and downregulation of MMP and ATP. However, in FSLLRY-NH2 (PAR2 antagonist) treated LO2 cells or PAR2-KO mice, PAR2 blockade reversed the above adverse effects of HBx on liver cells or tissues. CONCLUSION Inhibition of PAR2 may suppress inflammation and mitochondria oxidative stress caused by HBx, pointing out the potential application values of PAR2 antagonist on the treatment of HBV infection in clinic.
Collapse
Affiliation(s)
- Bin Li
- Laboratory of Immunology and Pathogenic Biology, Experimental Teaching Center of Basic Medicine, Jinzhou Medical University, Jinzhou City, Liaoning Province, 121001, People’s Republic of China
| | - Yonggang Li
- Department of Pathogenic Biology, School of Basic Medicine, Jinzhou Medical University, Jinzhou City, Liaoning Province, 121001, People’s Republic of China
| | - Shuhua Li
- Laboratory of Immunology and Pathogenic Biology, Experimental Teaching Center of Basic Medicine, Jinzhou Medical University, Jinzhou City, Liaoning Province, 121001, People’s Republic of China
| | - Hongwei Li
- Laboratory of Immunology and Pathogenic Biology, Experimental Teaching Center of Basic Medicine, Jinzhou Medical University, Jinzhou City, Liaoning Province, 121001, People’s Republic of China
| | - Ling Liu
- Laboratory of Immunology and Pathogenic Biology, Experimental Teaching Center of Basic Medicine, Jinzhou Medical University, Jinzhou City, Liaoning Province, 121001, People’s Republic of China
| | - Yao Xu
- School of Pharmacy, Jinzhou Medical University, Jinzhou City, Liaoning Province, 121001, People’s Republic of China
| |
Collapse
|
21
|
Michael E, Covic L, Kuliopulos A. Lipopeptide Pepducins as Therapeutic Agents. Methods Mol Biol 2021; 2383:307-333. [PMID: 34766299 DOI: 10.1007/978-1-0716-1752-6_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Pepducins are lipidated peptides that target the intracellular loops of G protein-coupled receptors (GPCRs) in order to modulate transmembrane signaling to internally located effectors. With a wide array of potential activities ranging from partial, biased, or full agonism to antagonism, pepducins represent a versatile class of compounds that can be used to potentially treat diverse human diseases or be employed as novel tools to probe complex mechanisms of receptor activation and signaling in cells and in animals. Here, we describe a number of different pepducins including an advanced compound, PZ-128, that has successfully progressed through phase 2 clinical trials in cardiac patients demonstrating safety and efficacy in suppressing myonecrosis and arterial thrombosis.
Collapse
Affiliation(s)
- Emily Michael
- Center of Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Lidija Covic
- Center of Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Athan Kuliopulos
- Center of Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
22
|
Jiang Y, Zhuo X, Wu Y, Fu X, Mao C. PAR2 blockade reverses osimertinib resistance in non-small-cell lung cancer cells via attenuating ERK-mediated EMT and PD-L1 expression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119144. [PMID: 34599981 DOI: 10.1016/j.bbamcr.2021.119144] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 08/28/2021] [Accepted: 09/09/2021] [Indexed: 12/25/2022]
Abstract
Osimertinib, as the third-generation EGFR tyrosine kinase inhibitors (EGFR-TKIs), is a first-line molecularly targeted drug for non-small cell lung cancer (NSCLC). However, the emergence of therapeutic resistance to osimertinib markedly impairs its efficiency and efficacy, leading to the failure of clinical applications. Novel molecular targets and drugs are urgently needed for reversing osimertinib resistance in NSCLC. Protease-activated receptor 2 (PAR2) that belongs to a subfamily of G protein-coupled receptors can stimulate the transactivation of EGFR to regulate multiple cellular signalling, actively participating in tumour progression. This study firstly discovered that PAR2 expression was notably enhanced when NSCLC cells became resistant to osimertinib. A PAR2 inhibitor facilitated osimertinib to attenuate EGFR transactivation, ERK phosphorylation, EMT and PD-L1 expression which were associated to osimertinib resistance. The combination of the PAR2 inhibitor and osimertinib also notably blocked cell viability, migration, 3D sphere formation and in vivo tumour growth whereas osimertinib itself lost such inhibitory effects in osimertinib-resistant NSCLC cells. Importantly, this reversal effect of PAR2 blockade was uncovered to depend on ERK-mediated EMT and PD-L1, since inhibition of β-arrestin or ERK, which could be modulated by PAR2, sensitized osimertinib to prevent EMT, PD-L1 expression and consequently overcame osimertinib resistance. Thus, this study demonstrated that PAR2 antagonism could limit ERK-mediated EMT and immune checkpoints, consequently attenuating EGFR transactivation and reactivate osimertinib. It suggested that PAR2 may be a novel drug target for osimertinib resistance, and PAR2 inhibition may be a promising strategy candidate for reversing EGFR-TKI resistance in NSCLC.
Collapse
Affiliation(s)
- Yuhong Jiang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, PR China.
| | - Xin Zhuo
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, PR China
| | - Yue Wu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, PR China
| | - Xiujuan Fu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, PR China
| | - Canquan Mao
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, PR China.
| |
Collapse
|
23
|
Ahmed IA, Jaffa MA, Moussa M, Hatem D, El-Achkar GA, Al Sayegh R, Karam M, Hamade E, Habib A, Jaffa AA. Plasma Kallikrein as a Modulator of Liver Injury/Remodeling. Front Pharmacol 2021; 12:715111. [PMID: 34566641 PMCID: PMC8458624 DOI: 10.3389/fphar.2021.715111] [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: 05/26/2021] [Accepted: 07/23/2021] [Indexed: 11/13/2022] Open
Abstract
The occurrence and persistence of hepatic injury which arises from cell death and inflammation result in liver disease. The processes that lead to liver injury progression and resolution are still not fully delineated. The plasma kallikrein-kinin system (PKKS) has been shown to play diverse functions in coagulation, tissue injury, and inflammation, but its role in liver injury has not been defined yet. In this study, we have characterized the role of the PKKS at various stages of liver injury in mice, as well as the direct effects of plasma kallikrein on human hepatocellular carcinoma cell line (HepG2). Histological, immunohistochemical, and gene expression analyses were utilized to assess cell injury on inflammatory and fibrotic factors. Acute liver injury triggered by carbon tetrachloride (CCl4) injection resulted in significant upregulation of the plasma kallikrein gene (Klkb1) and was highly associated with the high mobility group box 1 gene, the marker of cell death (r = 0.75, p < 0.0005, n = 7). In addition, increased protein expression of plasma kallikrein was observed as clusters around necrotic areas. Plasma kallikrein treatment significantly increased the proliferation of CCl4-induced HepG2 cells and induced a significant increase in the gene expression of the thrombin receptor (protease activated receptor-1), interleukin 1 beta, and lectin–galactose binding soluble 3 (galectin-3) (p < 0.05, n = 4). Temporal variations in the stages of liver fibrosis were associated with an increase in the mRNA levels of bradykinin receptors: beta 1 and 2 genes (p < 0.05; n = 3–10). In conclusion, these findings indicate that plasma kallikrein may play diverse roles in liver injury, inflammation, and fibrosis, and suggest that plasma kallikrein may be a target for intervention in the states of liver injury.
Collapse
Affiliation(s)
- Ibrahim A Ahmed
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, Beirut, Lebanon
| | - Miran A Jaffa
- Epidemiology and Population Health Department, Faculty of Health Sciences, American University of Beirut, Beirut, Lebanon
| | - Mayssam Moussa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, Beirut, Lebanon
| | - Duaa Hatem
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, Beirut, Lebanon.,Section of Pharmacology, Department of Bioethics and Safety, Catholic University, Rome, Italy
| | - Ghewa A El-Achkar
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, Beirut, Lebanon
| | - Rola Al Sayegh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, Beirut, Lebanon.,INSERM-UMR1149, Centre de Recherche sur l'Inflammation, and Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, Faculté de Médecine, Site Xavier Bichat, Universite de Paris, Paris, France
| | - Mia Karam
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, Beirut, Lebanon.,Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut, Lebanon
| | - Eva Hamade
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Beirut, Lebanon
| | - Aida Habib
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, Beirut, Lebanon.,INSERM-UMR1149, Centre de Recherche sur l'Inflammation, and Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, Faculté de Médecine, Site Xavier Bichat, Universite de Paris, Paris, France
| | - Ayad A Jaffa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, Beirut, Lebanon
| |
Collapse
|
24
|
The development of proteinase-activated receptor-2 modulators and the challenges involved. Biochem Soc Trans 2021; 48:2525-2537. [PMID: 33242065 PMCID: PMC7752072 DOI: 10.1042/bst20200191] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/13/2020] [Accepted: 11/02/2020] [Indexed: 11/30/2022]
Abstract
Protease-activated receptor-2 (PAR2) has been extensively studied since its discovery in the mid-1990. Despite the advances in understanding PAR2 pharmacology, it has taken almost 25 years for the first inhibitor to reach clinical trials, and so far, no PAR2 antagonist has been approved for human use. Research has employed classical approaches to develop a wide array of PAR2 agonists and antagonists, consisting of peptides, peptoids and antibodies to name a few, with a surge in patent applications over this period. Recent breakthroughs in PAR2 structure determination has provided a unique insight into proposed PAR2 ligand binding sites. Publication of the first crystal structures of PAR2 resolved in complex with two novel non-peptide small molecule antagonists (AZ8838 and AZ3451) revealed two distinct binding pockets, originally presumed to be allosteric sites, with a PAR2 antibody (Fab3949) used to block tethered ligand engagement with the peptide-binding domain of the receptor. Further studies have proposed orthosteric site occupancy for AZ8838 as a competitive antagonist. One company has taken the first PAR2 antibody (MEDI0618) into phase I clinical trial (NCT04198558). While this first-in-human trial is at the early stages of the assessment of safety, other research into the structural characterisation of PAR2 is still ongoing in an attempt to identify new ways to target receptor activity. This review will focus on the development of novel PAR2 modulators developed to date, with an emphasis placed upon the advances made in the pharmacological targeting of PAR2 activity as a strategy to limit chronic inflammatory disease.
Collapse
|
25
|
Kirolos SA, Rijal R, Consalvo KM, Gomer RH. Using Dictyostelium to Develop Therapeutics for Acute Respiratory Distress Syndrome. Front Cell Dev Biol 2021; 9:710005. [PMID: 34350188 PMCID: PMC8326840 DOI: 10.3389/fcell.2021.710005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) involves damage to lungs causing an influx of neutrophils from the blood into the lung airspaces, and the neutrophils causing further damage, which attracts more neutrophils in a vicious cycle. There are ∼190,000 cases of ARDS per year in the US, and because of the lack of therapeutics, the mortality rate is ∼40%. Repelling neutrophils out of the lung airspaces, or simply preventing neutrophil entry, is a potential therapeutic. In this minireview, we discuss how our lab noticed that a protein called AprA secreted by growing Dictyostelium cells functions as a repellent for Dictyostelium cells, causing cells to move away from a source of AprA. We then found that AprA has structural similarity to a human secreted protein called dipeptidyl peptidase IV (DPPIV), and that DPPIV is a repellent for human neutrophils. In animal models of ARDS, inhalation of DPPIV or DPPIV mimetics blocks neutrophil influx into the lungs. To move DPPIV or DPPIV mimetics into the clinic, we need to know how this repulsion works to understand possible drug interactions and side effects. Combining biochemistry and genetics in Dictyostelium to elucidate the AprA signal transduction pathway, followed by drug studies in human neutrophils to determine similarities and differences between neutrophil and Dictyostelium chemorepulsion, will hopefully lead to the safe use of DPPIV or DPPIV mimetics in the clinic.
Collapse
Affiliation(s)
| | | | | | - Richard H. Gomer
- Department of Biology, Texas A&M University, College Station, TX, United States
| |
Collapse
|
26
|
Kim BM, Kim DH, Park YJ, Ha S, Choi YJ, Yu HS, Chung KW, Chung HY. PAR2 promotes high-fat diet-induced hepatic steatosis by inhibiting AMPK-mediated autophagy. J Nutr Biochem 2021; 95:108769. [PMID: 34000413 DOI: 10.1016/j.jnutbio.2021.108769] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 02/24/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
Protease-activated receptor 2 (PAR2) is a member of G protein-coupled receptors. There are two types of PAR2 signaling pathways: Canonical G-protein signaling and β-arrestin signaling. Although PAR2 signaling has been reported to aggravate hepatic steatosis, the exact mechanism is still unclear, and the role of PAR2 in autophagy remains unknown. In this study, we investigated the regulatory role of PAR2 in autophagy during high-fat diet (HFD)-induced hepatic steatosis in mice. Increased protein levels of PAR2 and β-arrestin-2 and their interactions were detected after four months of HFD. To further investigate the role of PAR2, male and female wild-type (WT) and PAR2-knockout (PAR2 KO) mice were fed HFD. PAR2 deficiency protected HFD-induced hepatic steatosis in male mice, but not in female mice. Interestingly, PAR2-deficient liver showed increased AMP-activated protein kinase (AMPK) activation with decreased interaction between Ca2+/calmodulin-dependent protein kinase kinase β (CAMKKβ) and β-arrestin-2. In addition, PAR2 deficiency up-regulated autophagy in the liver. To elucidate whether PAR2 plays a role in the regulation of autophagy and lipid accumulation in vitro, PAR2 was overexpressed in HepG2 cells. Overexpression of PAR2 decreased AMPK activation with increased interaction of CAMKKβ with β-arrestin-2 and significantly inhibited autophagic responses in HepG2 cells. Inhibition of autophagy by PAR2 overexpression further exacerbated palmitate-induced lipid accumulation in HepG2 cells. Collectively, these findings suggest that the increase in the PAR2-β-arrestin-2-CAMKKβ complex by HFD inhibits AMPK-mediated autophagy, leading to the alleviation of hepatic steatosis.
Collapse
Affiliation(s)
- Byeong Moo Kim
- Department of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Dae Hyun Kim
- Department of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Yeo Jin Park
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea; Korean Convergence Medicine, University of Science and Technology, Daejeon 34054, Republic of Korea
| | - Sugyeong Ha
- Department of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Yeon Ja Choi
- Department of Biopharmaceutical Engineering, Division of Chemistry and Biotechnology, Dongguk University, Gyeongju, Republic of Korea
| | - Hak Sun Yu
- Department of Parasitology and Tropical Medicine, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Ki Wung Chung
- Department of Pharmacy, Pusan National University, Busan, Republic of Korea.
| | - Hae Young Chung
- Department of Pharmacy, Pusan National University, Busan, Republic of Korea.
| |
Collapse
|
27
|
Sufleţel RT, Melincovici CS, Gheban BA, Toader Z, Mihu CM. Hepatic stellate cells - from past till present: morphology, human markers, human cell lines, behavior in normal and liver pathology. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY 2021; 61:615-642. [PMID: 33817704 PMCID: PMC8112759 DOI: 10.47162/rjme.61.3.01] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Hepatic stellate cell (HSC), initially analyzed by von Kupffer, in 1876, revealed to be an extraordinary mesenchymal cell, essential for both hepatocellular function and lesions, being the hallmark of hepatic fibrogenesis and carcinogenesis. Apart from their implications in hepatic injury, HSCs play a vital role in liver development and regeneration, xenobiotic response, intermediate metabolism, and regulation of immune response. In this review, we discuss the current state of knowledge regarding HSCs morphology, human HSCs markers and human HSC cell lines. We also summarize the latest findings concerning their roles in normal and liver pathology, focusing on their impact in fibrogenesis, chronic viral hepatitis and liver tumors.
Collapse
Affiliation(s)
- Rada Teodora Sufleţel
- Discipline of Histology, Department of Morphological Sciences, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania;
| | | | | | | | | |
Collapse
|
28
|
Kim DH, Kim YR, Bang E, Ha S, Noh SG, Kim BM, Jeong SH, Jung HJ, Lee JY, Chung HY. Mechanism of Lipid Accumulation through PAR2 Signaling in Diabetic Male Mice. Endocrinol Metab (Seoul) 2021; 36:171-184. [PMID: 33677938 PMCID: PMC7937841 DOI: 10.3803/enm.2020.850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/04/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Protease-activated protein-2 (PAR2) has been reported to regulate hepatic insulin resistance condition in type 2 diabetes mice. However, the mechanism of lipid metabolism through PAR2 in obesity mice have not yet been examined. In liver, Forkhead box O1 (FoxO1) activity induces peroxisome proliferator-activated receptor γ (PPARγ), leading to accumulation of lipids and hyperlipidemia. Hyperlipidemia significantly influence hepatic steatoses, but the mechanisms underlying PAR2 signaling are complex and have not yet been elucidated. METHODS To examine the modulatory action of FoxO1 and its altered interaction with PPARγ, we utilized db/db mice and PAR2-knockout (KO) mice administered with high-fat diet (HFD). RESULTS Here, we demonstrated that PAR2 was overexpressed and regulated downstream gene expressions in db/db but not in db+ mice. The interaction between PAR2/β-arrestin and Akt was also greater in db/db mice. The Akt inhibition increased FoxO1 activity and subsequently PPARγ gene in the livers that led to hepatic lipid accumulation. Our data showed that FoxO1 was negatively controlled by Akt signaling and consequently, the activity of a major lipogenesis-associated transcription factors such as PPARγ increased, leading to hepatic lipid accumulation through the PAR2 pathway under hyperglycemic conditions in mice. Furthermore, the association between PPARγ and FoxO1 was increased in hepatic steatosis condition in db/db mice. However, HFD-fed PAR2-KO mice showed suppressed FoxO1-induced hepatic lipid accumulation compared with HFD-fed control groups. CONCLUSION Collectively, our results provide evidence that the interaction of FoxO1 with PPARγ promotes hepatic steatosis in mice. This might be due to defects in PAR2/β-arrestin-mediated Akt signaling in diabetic and HFD-fed mice.
Collapse
Affiliation(s)
- Dae Hyun Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Korea
| | - Ye Ra Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Korea
| | - EunJin Bang
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Korea
| | - Sugyeong Ha
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Korea
| | - Sang Gyun Noh
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Korea
| | - Byeong Moo Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Korea
| | - Seong Ho Jeong
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Korea
| | - Hee Jin Jung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Korea
| | - Ji Young Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Korea
| | - Hae Young Chung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Korea
| |
Collapse
|
29
|
Healing gone wrong: convergence of hemostatic pathways and liver fibrosis? Clin Sci (Lond) 2020; 134:2189-2201. [PMID: 32844997 DOI: 10.1042/cs20191102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 08/16/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022]
Abstract
Fibrosis results from a disordered wound healing response within the liver with activated hepatic stellate cells laying down dense, collagen-rich extracellular matrix that eventually restricts liver hepatic synthetic function and causes increased sinusoidal resistance. The end result of progressive fibrosis, cirrhosis, is associated with significant morbidity and mortality as well as tremendous economic burden. Fibrosis can be conceptualized as an aberrant wound healing response analogous to a chronic ankle sprain that is driven by chronic liver injury commonly over decades. Two unique aspects of hepatic fibrosis - the chronic nature of insult required and the liver's unique ability to regenerate - give an opportunity for pharmacologic intervention to stop or slow the pace of fibrosis in patients early in the course of their liver disease. Two potential biologic mechanisms link together hemostasis and fibrosis: focal parenchymal extinction and direct stellate cell activation by thrombin and Factor Xa. Available translational research further supports the role of thrombosis in fibrosis. In this review, we will summarize what is known about the convergence of hemostatic changes and hepatic fibrosis in chronic liver disease and present current preclinical and clinical data exploring the relationship between the two. We will also present clinical trial data that underscores the potential use of anticoagulant therapy as an antifibrotic factor in liver disease.
Collapse
|
30
|
Rehman A, Baloch NUA, Morrow JP, Pacher P, Haskó G. Targeting of G-protein coupled receptors in sepsis. Pharmacol Ther 2020; 211:107529. [PMID: 32197794 PMCID: PMC7388546 DOI: 10.1016/j.pharmthera.2020.107529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/11/2022]
Abstract
The Third International Consensus Definitions (Sepsis-3) define sepsis as life-threatening multi-organ dysfunction caused by a dysregulated host response to infection. Sepsis can progress to septic shock-an even more lethal condition associated with profound circulatory, cellular and metabolic abnormalities. Septic shock remains a leading cause of death in intensive care units and carries a mortality of almost 25%. Despite significant advances in our understanding of the pathobiology of sepsis, therapeutic interventions have not translated into tangible differences in the overall outcome for patients. Clinical trials of antagonists of various pro-inflammatory mediators in sepsis have been largely unsuccessful in the past. Given the diverse physiologic roles played by G-protein coupled receptors (GPCR), modulation of GPCR signaling for the treatment of sepsis has also been explored. Traditional pharmacologic approaches have mainly focused on ligands targeting the extracellular domains of GPCR. However, novel techniques aimed at modulating GPCR intracellularly through aptamers, pepducins and intrabodies have opened a fresh avenue of therapeutic possibilities. In this review, we summarize the diverse roles played by various subfamilies of GPCR in the pathogenesis of sepsis and identify potential targets for pharmacotherapy through these novel approaches.
Collapse
Affiliation(s)
- Abdul Rehman
- Department of Medicine, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Noor Ul-Ain Baloch
- Department of Medicine, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - John P Morrow
- Department of Medicine, Columbia University, New York City, NY, United States
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York City, NY, United States.
| |
Collapse
|
31
|
Abstract
As basic research into GPCR signaling and its association with disease has come into fruition, greater clarity has emerged with regards to how these receptors may be amenable to therapeutic intervention. As a diverse group of receptor proteins, which regulate a variety of intracellular signaling pathways, research in this area has been slow to yield tangible therapeutic agents for the treatment of a number of diseases including cancer. However, recently such research has gained momentum based on a series of studies that have sought to define GPCR proteins dynamics through the elucidation of their crystal structures. In this chapter, we define the approaches that have been adopted in developing better therapeutics directed against the specific parts of the receptor proteins, such as the extracellular and the intracellular domains, including the ligands and auxiliary proteins that bind them. Finally, we also briefly outline how GPCR-derived signaling transduction pathways hold great potential as additional targets.
Collapse
Affiliation(s)
- Surinder M Soond
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation.
| |
Collapse
|
32
|
Weiskirchen R, Meurer SK, Liedtke C, Huber M. Mast Cells in Liver Fibrogenesis. Cells 2019; 8:E1429. [PMID: 31766207 PMCID: PMC6912398 DOI: 10.3390/cells8111429] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/05/2019] [Accepted: 11/10/2019] [Indexed: 01/10/2023] Open
Abstract
Mast cells (MCs) are immune cells of the myeloid lineage that are present in the connective tissue throughout the body and in mucosa tissue. They originate from hematopoietic stem cells in the bone marrow and circulate as MC progenitors in the blood. After migration to various tissues, they differentiate into their mature form, which is characterized by a phenotype containing large granules enriched in a variety of bioactive compounds, including histamine and heparin. These cells can be activated in a receptor-dependent and -independent manner. Particularly, the activation of the high-affinity immunoglobulin E (IgE) receptor, also known as FcεRI, that is expressed on the surface of MCs provoke specific signaling cascades that leads to intracellular calcium influx, activation of different transcription factors, degranulation, and cytokine production. Therefore, MCs modulate many aspects in physiological and pathological conditions, including wound healing, defense against pathogens, immune tolerance, allergy, anaphylaxis, autoimmune defects, inflammation, and infectious and other disorders. In the liver, MCs are mainly associated with connective tissue located in the surrounding of the hepatic arteries, veins, and bile ducts. Recent work has demonstrated a significant increase in MC number during hepatic injury, suggesting an important role of these cells in liver disease and progression. In the present review, we summarize aspects of MC function and mediators in experimental liver injury, their interaction with other hepatic cell types, and their contribution to the pathogenesis of fibrosis.
Collapse
Affiliation(s)
- Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), University Hospital, RWTH Aachen University, D-52074 Aachen, Germany;
| | - Steffen K. Meurer
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), University Hospital, RWTH Aachen University, D-52074 Aachen, Germany;
| | - Christian Liedtke
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany;
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, D-52074 Aachen, Germany
| |
Collapse
|
33
|
Rana R, Shearer AM, Fletcher EK, Nguyen N, Guha S, Cox DH, Abdelmalek M, Wang Y, Baleja JD, Covic L, Kuliopulos A. PAR2 controls cholesterol homeostasis and lipid metabolism in nonalcoholic fatty liver disease. Mol Metab 2019; 29:99-113. [PMID: 31668396 PMCID: PMC6742970 DOI: 10.1016/j.molmet.2019.08.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/16/2019] [Accepted: 08/24/2019] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE Increases in hepatic and plasma cholesterol occur in patients with nonalcoholic fatty liver disease (NAFLD), although the reason for this is not well understood. We investigated whether Protease-Activated Receptor 2 (PAR2) plays a role in cholesterol and lipid homeostasis in NAFLD. METHODS Human liver biopsies (n = 108) were quantified for PAR2 expression from NAFLD cases randomly selected and stratified by liver fibrosis stage, the primary predictor for clinical outcomes, while controlling for age, gender, and BMI between fibrosis groups. Demographic data and laboratory studies on plasma samples were obtained within 6 months of liver biopsy. Wild-type and PAR2-KO (C57BL/6 F2rl1-/-) mice were fed either normal or high fat diet for 16 weeks and plasma and liver assayed for lipids and soluble metabolites. RESULTS Severity of NAFLD and plasma cholesterol levels significantly correlated with hepatocyte PAR2 expression in NAFLD patients. Conversely, PAR2 deficiency in mice resulted in reduced expression of key hepatic genes involved in cholesterol synthesis, a 50% drop in plasma and total liver cholesterol, and induced a reverse cholesterol transport system that culminated in 25% higher fecal bile acid output. PAR2-deficient mice exhibited enhanced fatty acid β-oxidation with a ketogenic shift and an unexpected increase in liver glycogenesis. Mechanistic studies identified Gi-Jnk1/2 as key downstream effectors of protease-activated PAR2 in the regulation of lipid and cholesterol homeostasis in liver. CONCLUSIONS These data indicate that PAR2 may be a new target for the suppression of plasma cholesterol and hepatic fat accumulation in NAFLD and related metabolic conditions.
Collapse
Affiliation(s)
- Rajashree Rana
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA
| | - Andrew M Shearer
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA; Tufts University School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Elizabeth K Fletcher
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA
| | - Nga Nguyen
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA
| | - Srijoy Guha
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA
| | - Daniel H Cox
- Tufts University School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Manal Abdelmalek
- Division of Gastroenterology and Hepatology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Ying Wang
- Division of Gastroenterology and Hepatology, Duke University Medical Center, Durham, NC, 27710, USA
| | - James D Baleja
- Tufts University School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Lidija Covic
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA; Tufts University School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Athan Kuliopulos
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA; Tufts University School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA.
| |
Collapse
|
34
|
Kostallari E, Shah VH. Pericytes in the Liver. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1122:153-167. [PMID: 30937868 DOI: 10.1007/978-3-030-11093-2_9] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Liver pericytes, commonly named hepatic stellate cells (HSCs), reside in the space between liver sinusoidal endothelial cells (LSECs) and hepatocytes. They display important roles in health and disease. HSCs ensure the storage of the majority of vitamin A in a healthy body, and they represent the major source of fibrotic tissue in liver disease. Surrounding cells, such as LSECs, hepatocytes, and Kupffer cells, present a significant role in modulating HSC behavior. Therapeutic strategies against liver disease are being currently developed, where HSCs represent an ideal target. In this chapter, we will discuss HSC quiescence and activation in the context of healthy liver and diseases, such as fibrosis, steatohepatitis, and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Enis Kostallari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
35
|
Poujol de Molliens M, Jamadagni P, Létourneau M, Devost D, Hébert TE, Patten SA, Fournier A, Chatenet D. Design and biological assessment of membrane-tethering neuroprotective peptides derived from the pituitary adenylate cyclase-activating polypeptide type 1 receptor. Biochim Biophys Acta Gen Subj 2019; 1863:129398. [PMID: 31306709 DOI: 10.1016/j.bbagen.2019.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/03/2019] [Accepted: 07/10/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND The pituitary adenylate cyclase-activating polypeptide (PACAP) type 1 receptor (PAC1), a class B G protein-coupled receptor (GPCR), has emerged as a promising target for treating neurodegenerative conditions. Unfortunately, despite years of research, no PAC1-specific agonist has been discovered, as activity on two other GPCRs, VPAC1 and VPAC2, is retained with current analogs. Cell signaling is related to structural modifications in the intracellular loops (ICLs) of GPCRs. Thus, we hypothesized that peptides derived from the ICLs (called pepducins) of PAC1 might initiate, as allosteric ligands, signaling cascades after recognition of the parent receptor and modulation of its conformational landscape. METHODS Three pepducins were synthesized and evaluated for their ability to 1) promote cell survival; 2) stimulate various signaling pathways associated with PAC1 activation; 3) modulate selectively PAC1, VPAC1 or VPAC2 activation; and 4) sustain mobility and prevent death of dopaminergic neurons in a zebrafish model of neurodegeneration. RESULTS Assays demonstrated that these molecules promote SH-SY5Y cell survival, a human neuroblastoma cell line expressing PAC1, and activate signaling via Gαs and Gαq, with distinct potencies and efficacies. Also, PAC1-Pep1 and PAC1-Pep2 activated selectively PAC1-mediated Gαs stimulation. Finally, experiments, using a zebrafish neurodegeneration model, showed a neuroprotective action with all three pepducins and in particular, revealed the ability of PAC1-Pep1 and PAC1-Pep3 to preserve fish mobility and tyrosine hydroxylase expression in the brain. CONCLUSION We have developed the first neuroprotective pepducins derived from PAC1, a class B GPCR. GENERAL SIGNIFICANCE PAC1-derived pepducins represent attractive templates for the development of innovative neuroprotecting molecules.
Collapse
Affiliation(s)
- Mathilde Poujol de Molliens
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, QC, Canada; Institut National de la Recherche Scientifique, Centre Armand-Frappier, Laboratoire d'études moléculaires et pharmacologiques des peptides (LEMPP), Université du Québec, Ville de Laval, QC, Canada
| | - Priyanka Jamadagni
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Université du Québec, Ville de Laval, QC, Canada
| | - Myriam Létourneau
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, QC, Canada; Institut National de la Recherche Scientifique, Centre Armand-Frappier, Laboratoire d'études moléculaires et pharmacologiques des peptides (LEMPP), Université du Québec, Ville de Laval, QC, Canada
| | - Dominic Devost
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Shunmoogum A Patten
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Université du Québec, Ville de Laval, QC, Canada
| | - Alain Fournier
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Laboratoire d'études moléculaires et pharmacologiques des peptides (LEMPP), Université du Québec, Ville de Laval, QC, Canada
| | - David Chatenet
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, QC, Canada.
| |
Collapse
|
36
|
Kim DH, Lee B, Lee J, Kim ME, Lee JS, Chung JH, Yu BP, Dong HH, Chung HY. FoxO6-mediated IL-1β induces hepatic insulin resistance and age-related inflammation via the TF/PAR2 pathway in aging and diabetic mice. Redox Biol 2019; 24:101184. [PMID: 30974318 PMCID: PMC6454229 DOI: 10.1016/j.redox.2019.101184] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 03/18/2019] [Accepted: 03/28/2019] [Indexed: 12/24/2022] Open
Abstract
FoxO has been proposed to play a role in the promotion of insulin resistance, and inflammation. FoxO is a pro-inflammatory transcription factor that is a key mediator of generation of inflammatory cytokines such as IL-1β in the liver. However, the detailed association of FoxO6 with insulin resistance and age-related inflammation has not been fully documented. Here, we showed that FoxO6 was elevated in the livers of aging rats and obese mice that exhibited insulin resistance. In addition, virus-mediated FoxO6 activation led to insulin resistance in mice with a notable increase in PAR2 and inflammatory signaling in the liver. On the other hand, FoxO6-KO mice showed reduced PAR2 signaling with a decrease in inflammatory cytokine expression and elevated insulin signaling. Because FoxO6 is closely associated with abnormal production of IL-1β in the liver, we focused on the FoxO6/IL-1β/PAR2 axis to further examine mechanisms underlying FoxO6-mediated insulin resistance and inflammation in the liver. In vitro experiments showed that FoxO6 directly binds to and elevates IL-1β expression. In turn, IL-1β treatment elevated the protein levels of PAR2 with a significant decrease in hepatic insulin signaling, whereas PAR2-siRNA treatment abolished these effects. However, PAR2-siRNA treatment had no effect on IL-1β expression induced by FoxO6, indicating that IL-1β may not be downstream of PAR2. Taken together, we assume that FoxO6-mediated IL-1β is involved in hepatic inflammation and insulin resistance via TF/PAR2 pathway in the liver.
Collapse
Affiliation(s)
- Dae Hyun Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2, Busandaehak-ro 63beon-gi, Geumjeong-Gu, Busan, 46241, South Korea
| | - Bonggi Lee
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu, 41062, Republic of Korea
| | - Jaewon Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2, Busandaehak-ro 63beon-gi, Geumjeong-Gu, Busan, 46241, South Korea
| | - Mi Eun Kim
- Department of Biology, College of Natural Science, Chosun University, Gwangju, 501-759, Republic of Korea
| | - Jun Sik Lee
- Department of Biology, College of Natural Science, Chosun University, Gwangju, 501-759, Republic of Korea
| | - Jae Heun Chung
- Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, 50612, South Korea
| | - Byung Pal Yu
- Department of Physiology, The University of Texas Health Science Center at San Antonio, TX, 78229, USA
| | - H Henry Dong
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15224, USA
| | - Hae Young Chung
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2, Busandaehak-ro 63beon-gi, Geumjeong-Gu, Busan, 46241, South Korea.
| |
Collapse
|
37
|
Lee MS, Lerner EA. Targeting PAR2 with Pepducins. J Invest Dermatol 2019; 139:282-284. [DOI: 10.1016/j.jid.2018.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 09/18/2018] [Accepted: 09/18/2018] [Indexed: 11/24/2022]
|
38
|
Michael ES, Covic L, Kuliopulos A. Trace amine-associated receptor 1 (TAAR1) promotes anti-diabetic signaling in insulin-secreting cells. J Biol Chem 2019; 294:4401-4411. [PMID: 30670596 DOI: 10.1074/jbc.ra118.005464] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 01/16/2019] [Indexed: 12/25/2022] Open
Abstract
Pancreatic β-cell failure in type 2 diabetes mellitus is a serious challenge that results in an inability of the pancreas to produce sufficient insulin to properly regulate blood glucose levels. Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor expressed by β-cells that has recently been proposed as a potential target for improving glycemic control and suppressing binge eating behaviors. We discovered that TAAR1 is coupled to Gαs-signaling pathways in insulin-secreting β-cells to cause protein kinase A (PKA)/exchange protein activated by cAMP (Epac)-dependent release of insulin, activation of RAF proto-oncogene, Ser/Thr kinase (Raf)-mitogen-activated protein kinase (MAPK) signaling, induction of cAMP response element-binding protein (CREB)-insulin receptor substrate 2 (Irs-2), and increased β-cell proliferation. Interestingly, TAAR1 triggered cAMP-mediated calcium influx and release from internal stores, both of which were required for activation of a MAPK cascade utilizing calmodulin-dependent protein kinase II (CaMKII), Raf, and MAPK/ERK kinase 1/2 (MEK1/2). Together, these data identify TAAR1/Gαs-mediated signaling pathways that promote insulin secretion, improved β-cell function and proliferation, and highlight TAAR1 as a promising new target for improving β-cell health in type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Emily S Michael
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Graduate Biomedical Sciences/Biochemistry, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Lidija Covic
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Graduate Biomedical Sciences/Biochemistry, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Athan Kuliopulos
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Graduate Biomedical Sciences/Biochemistry, Tufts University School of Medicine, Boston, Massachusetts 02111
| |
Collapse
|
39
|
Zhao Q, Xu H, Hong S, Song N, Xie J, Yan Z, Wang R, Yang P, Jiang X. Rapeseed Protein-Derived Antioxidant Peptide RAP Ameliorates Nonalcoholic Steatohepatitis and Related Metabolic Disorders in Mice. Mol Pharm 2018; 16:371-381. [PMID: 30543441 DOI: 10.1021/acs.molpharmaceut.8b01030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Rapeseed protein hydrolysates have recently shown in vitro antioxidant and anti-inflammatory activities. However, scant data exist about their in vivo activities. Here, we report that the peptide DHNNPQIR (hereinafter referred to as RAP-8), a bioactive peptide originated from rapeseed protein, exhibits excellent in vivo efficacy in mouse models of nonalcoholic steatohepatitis (NASH) and hepatic fibrosis. We demonstrated that RAP-8 significantly reduced hepatic steatosis and improved insulin resistance and lipid metabolism. Furthermore, RAP-8 showed markedly reduced hepatic inflammation, fibrosis, liver injury, and metabolic deterioration. In particular, RAP-8 directly suppressed fibrosis-associated gene expression, including α-smooth muscle actin (α-Sma) and collagen type I (Col-1α) in the liver of mice in vivo. In addtion, RAP-8 significantly decreased macrophage infiltration and reduced pro-inflammatory cytokines secretion. Finally, we found that RAP-8 administration significantly decreased oxidative stress-induced apoptosis in liver injury induced by CCl4. Therefore, our results suggest that RAP-8 could be available for treatment of NASH and NASH-related metabolic disorders as a potential therapeutic candidate.
Collapse
Affiliation(s)
- Qian Zhao
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510000 , China
| | - Hongjiao Xu
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510000 , China
| | - Sihua Hong
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510000 , China
| | - Nazi Song
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510000 , China
| | - Junqiu Xie
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510000 , China.,Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences , Lanzhou University , Lanzhou , China
| | - Zhibin Yan
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences , Lanzhou University , Lanzhou , China
| | - Rui Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences , Lanzhou University , Lanzhou , China
| | - Pengyu Yang
- Innopep Inc. , San Diego , California 92121 , United States
| | - Xianxing Jiang
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510000 , China
| |
Collapse
|
40
|
Mastocytosis-derived extracellular vesicles exhibit a mast cell signature, transfer KIT to stellate cells, and promote their activation. Proc Natl Acad Sci U S A 2018; 115:E10692-E10701. [PMID: 30352845 DOI: 10.1073/pnas.1809938115] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) have been implicated in the development and progression of hematological malignancies. We thus examined serum samples from patients with systemic mastocytosis (SM) and found EVs with a mast cell signature including the presence of tryptase, FcεRI, MRGX2, and KIT. The concentration of these EVs correlated with parameters of disease including levels of serum tryptase, IL-6, and alkaline phosphatase and physical findings including hepatosplenomegaly. Given reports that EVs from one cell type may influence another cell's behavior, we asked whether SM-EVs might affect hepatic stellate cells (HSCs), based on the abnormal liver pathology associated with mastocytosis. We found that KIT was transferred from SM-EVs into an HSC line eliciting proliferation, cytokine production, and differentiation, processes that have been associated with liver pathology. These effects were reduced by KIT inhibition or neutralization and recapitulated by enforced expression of KIT or constitutively active D816V-KIT, a gain-of-function variant associated with SM. Furthermore, HSCs in liver from mice injected with SM-EVs had increased expression of α-SMA and human KIT, particularly around portal areas, compared with mice injected with EVs from normal individuals, suggesting that SM-EVs can also initiate HSC activation in vivo. Our data are thus consistent with the conclusion that SM-EVs have the potential to influence cells outside the hematological compartment and that therapeutic approaches for treatment of SM may be effective in part through inhibition of effects of EVs on target tissues, findings important both to understanding complex disease pathology and in developing interventional agents for the treatment of hematologic diseases.
Collapse
|
41
|
Barr TP, Garzia C, Guha S, Fletcher EK, Nguyen N, Wieschhaus AJ, Ferrer L, Covic L, Kuliopulos A. PAR2 Pepducin-Based Suppression of Inflammation and Itch in Atopic Dermatitis Models. J Invest Dermatol 2018; 139:412-421. [PMID: 30287285 DOI: 10.1016/j.jid.2018.08.019] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/09/2018] [Accepted: 08/12/2018] [Indexed: 02/06/2023]
Abstract
PAR2 has been proposed to contribute to lesion formation and intense itch in atopic dermatitis. Here, we tested the ability of a cell-penetrating pepducin, PZ-235, to mitigate the potentially deleterious effects of PAR2 in models of atopic dermatitis. PZ-235 significantly inhibited PAR2-mediated expression of inflammatory factors NF-κB, TSLP, TNF-α, and differentiation marker K10 by 94%-98% (P < 0.001) in human keratinocytes and suppressed IL-4 and IL-13 by 68%-83% (P < 0.05) in mast cells. In delayed pepducin treatment models of oxazolone- and DNFB-induced dermatitis, PZ-235 significantly attenuated skin thickening by 43%-100% (P < 0.01) and leukocyte crusting by 57% (P < 0.05), and it inhibited ex vivo chemotaxis of leukocytes toward PAR2 agonists. Daily PZ-235 treatment of filaggrin-deficient mice exposed to dust mite allergens for 8 weeks significantly suppressed total leukocyte and T-cell infiltration by 50%-68%; epidermal thickness by 60%-77%; and skin thickening, scaling, excoriation, and total lesion severity score by 46%-56%. PZ-235 significantly reduced itching caused by wasp venom peptide degranulation of mast cells in mice by 51% (P < 0.05), which was comparable to the protective effects conferred by PAR2 deficiency. Taken together, these results suggest that a PAR2 pepducin may confer broad therapeutic benefits as a disease-modifying treatment for atopic dermatitis and itch.
Collapse
Affiliation(s)
- Travis P Barr
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Chris Garzia
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Srijoy Guha
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Elizabeth K Fletcher
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Nga Nguyen
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Adam J Wieschhaus
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Lluis Ferrer
- Department of Dermatology, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, USA
| | - Lidija Covic
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Athan Kuliopulos
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA.
| |
Collapse
|
42
|
Grisanti LA, Thomas TP, Carter RL, de Lucia C, Gao E, Koch WJ, Benovic JL, Tilley DG. Pepducin-mediated cardioprotection via β-arrestin-biased β2-adrenergic receptor-specific signaling. Theranostics 2018; 8:4664-4678. [PMID: 30279730 PMCID: PMC6160776 DOI: 10.7150/thno.26619] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/21/2018] [Indexed: 12/20/2022] Open
Abstract
Reperfusion as a therapeutic intervention for acute myocardial infarction-induced cardiac injury itself induces further cardiomyocyte death. β-arrestin (βarr)-biased β-adrenergic receptor (βAR) activation promotes survival signaling responses in vitro; thus, we hypothesize that this pathway can mitigate cardiomyocyte death at the time of reperfusion to better preserve function. However, a lack of efficacious βarr-biased orthosteric small molecules has prevented investigation into whether this pathway relays protection against ischemic injury in vivo. We recently demonstrated that the pepducin ICL1-9, a small lipidated peptide fragment designed from the first intracellular loop of β2AR, allosterically engaged pro-survival signaling cascades in a βarr-dependent manner in vitro. Thus, in this study we tested whether ICL1-9 relays cardioprotection against ischemia/reperfusion (I/R)-induced injury in vivo. Methods: Wild-type (WT) C57BL/6, β2AR knockout (KO), βarr1KO and βarr2KO mice received intracardiac injections of either ICL1-9 or a scrambled control pepducin (Scr) at the time of ischemia (30 min) followed by reperfusion for either 24 h, to assess infarct size and cardiomyocyte death, or 4 weeks, to monitor the impact of ICL1-9 on long-term cardiac structure and function. Neonatal rat ventricular myocytes (NRVM) were used to assess the impact of ICL1-9 versus Scr pepducin on cardiomyocyte survival and mitochondrial superoxide formation in response to either serum deprivation or hypoxia/reoxygenation (H/R) in vitro and to investigate the associated mechanism(s). Results: Intramyocardial injection of ICL1-9 at the time of I/R reduced infarct size, cardiomyocyte death and improved cardiac function in a β2AR- and βarr-dependent manner, which led to improved contractile function early and less fibrotic remodeling over time. Mechanistically, ICL1-9 attenuated mitochondrial superoxide production and promoted cardiomyocyte survival in a RhoA/ROCK-dependent manner. RhoA activation could be detected in cardiomyocytes and whole heart up to 24 h post-treatment, demonstrating the stability of ICL1-9 effects on βarr-dependent β2AR signaling. Conclusion: Pepducin-based allosteric modulation of βarr-dependent β2AR signaling represents a novel therapeutic approach to reduce reperfusion-induced cardiac injury and relay long-term cardiac remodeling benefits.
Collapse
|
43
|
Pant A, Kopec AK, Luyendyk JP. Role of the blood coagulation cascade in hepatic fibrosis. Am J Physiol Gastrointest Liver Physiol 2018; 315:G171-G176. [PMID: 29723040 PMCID: PMC6139645 DOI: 10.1152/ajpgi.00402.2017] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/19/2018] [Accepted: 04/24/2018] [Indexed: 02/07/2023]
Abstract
Liver is the primary source of numerous proteins that are critical for normal function of the blood coagulation cascade. Because of this, diseases of the liver, particularly when affiliated with severe complications like cirrhosis, are associated with abnormalities of blood clotting. Although conventional interpretation has inferred cirrhosis as a disorder of uniform bleeding risk, it is now increasingly appreciated as a disease wherein the coagulation cascade is precariously rebalanced. Moreover, prothrombotic risk factors are also associated with a more rapid progression of fibrosis in humans, suggesting that coagulation proteases participate in disease pathogenesis. Indeed, strong evidence drawn from experimental animal studies indicates that components of the coagulation cascade, particularly coagulation factor Xa and thrombin, drive profibrogenic events, leading to hepatic fibrosis. Here, we concisely review the evidence supporting a pathologic role for coagulation in the development of liver fibrosis and the potential mechanisms involved. Further, we highlight how studies in experimental animals may shed light on emerging clinical evidence, suggesting that beneficial effects of anticoagulation could extend beyond preventing thrombotic complications to include reducing pathologies like fibrosis.
Collapse
Affiliation(s)
- Asmita Pant
- Department of Pathobiology and Diagnostic Investigation, Michigan State University , East Lansing, Michigan
- Institute for Integrative Toxicology, Michigan State University , East Lansing, Michigan
| | - Anna K Kopec
- Department of Pathobiology and Diagnostic Investigation, Michigan State University , East Lansing, Michigan
- Institute for Integrative Toxicology, Michigan State University , East Lansing, Michigan
| | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University , East Lansing, Michigan
- Institute for Integrative Toxicology, Michigan State University , East Lansing, Michigan
- Department of Pharmacology and Toxicology, Michigan State University , East Lansing, Michigan
| |
Collapse
|
44
|
Signaling Crosstalk of TGF-β/ALK5 and PAR2/PAR1: A Complex Regulatory Network Controlling Fibrosis and Cancer. Int J Mol Sci 2018; 19:ijms19061568. [PMID: 29795022 PMCID: PMC6032192 DOI: 10.3390/ijms19061568] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/09/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023] Open
Abstract
Both signaling by transforming growth factor-β (TGF-β) and agonists of the G Protein-coupled receptors proteinase-activated receptor-1 (PAR1) and -2 (PAR2) have been linked to tissue fibrosis and cancer. Intriguingly, TGF-β and PAR signaling either converge on the regulation of certain matrix genes overexpressed in these pathologies or display mutual regulation of their signaling components, which is mediated in part through sphingosine kinases and sphingosine-1-phosphate and indicative of an intimate signaling crosstalk between the two pathways. In the first part of this review, we summarize the various regulatory interactions that have been discovered so far according to the organ/tissue in which they were described. In the second part, we highlight the types of signaling crosstalk between TGF-β on the one hand and PAR2/PAR1 on the other hand. Both ligand–receptor systems interact at various levels and by several mechanisms including mutual regulation of ligand–ligand, ligand–receptor, and receptor–receptor at the transcriptional, post-transcriptional, and receptor transactivation levels. These mutual interactions between PAR2/PAR1 and TGF-β signaling components eventually result in feed-forward loops/vicious cycles of matrix deposition and malignant traits that exacerbate fibrosis and oncogenesis, respectively. Given the crucial role of PAR2 and PAR1 in controlling TGF-β receptor activation, signaling, TGF-β synthesis and bioactivation, combining PAR inhibitors with TGF-β blocking agents may turn out to be more efficient than targeting TGF-β alone in alleviating unwanted TGF-β-dependent responses but retaining the beneficial ones.
Collapse
|
45
|
Chaturvedi M, Schilling J, Beautrait A, Bouvier M, Benovic JL, Shukla AK. Emerging Paradigm of Intracellular Targeting of G Protein-Coupled Receptors. Trends Biochem Sci 2018; 43:533-546. [PMID: 29735399 DOI: 10.1016/j.tibs.2018.04.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 01/12/2023]
Abstract
G protein-coupled receptors (GPCRs) recognize a diverse array of extracellular stimuli, and they mediate a broad repertoire of signaling events involved in human physiology. Although the major effort on targeting GPCRs has typically been focused on their extracellular surface, a series of recent developments now unfold the possibility of targeting them from the intracellular side as well. Allosteric modulators binding to the cytoplasmic surface of GPCRs have now been described, and their structural mechanisms are elucidated by high-resolution crystal structures. Furthermore, pepducins, aptamers, and intrabodies targeting the intracellular face of GPCRs have also been successfully utilized to modulate receptor signaling. Moreover, small molecule compounds, aptamers, and synthetic intrabodies targeting β-arrestins have also been discovered to modulate GPCR endocytosis and signaling. Here, we discuss the emerging paradigm of intracellular targeting of GPCRs, and outline the current challenges, potential opportunities, and future outlook in this particular area of GPCR biology.
Collapse
Affiliation(s)
- Madhu Chaturvedi
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Justin Schilling
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Alexandre Beautrait
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, H3T 1J4, Canada
| | - Michel Bouvier
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, H3T 1J4, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Quebec, H3T 1J4, Canada
| | - Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Arun K Shukla
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India.
| |
Collapse
|
46
|
Rana R, Huang T, Koukos G, Fletcher EK, Turner SE, Shearer A, Gurbel PA, Rade JJ, Kimmelstiel CD, Bliden KP, Covic L, Kuliopulos A. Noncanonical Matrix Metalloprotease 1-Protease-Activated Receptor 1 Signaling Drives Progression of Atherosclerosis. Arterioscler Thromb Vasc Biol 2018; 38:1368-1380. [PMID: 29622563 DOI: 10.1161/atvbaha.118.310967] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/22/2018] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Protease-activated receptor-1 (PAR1) is classically activated by thrombin and is critical in controlling the balance of hemostasis and thrombosis. More recently, it has been shown that noncanonical activation of PAR1 by matrix metalloprotease-1 (MMP1) contributes to arterial thrombosis. However, the role of PAR1 in long-term development of atherosclerosis is unknown, regardless of the protease agonist. APPROACH AND RESULTS We found that plasma MMP1 was significantly correlated (R=0.33; P=0.0015) with coronary atherosclerotic burden as determined by angiography in 91 patients with coronary artery disease and acute coronary syndrome undergoing cardiac catheterization or percutaneous coronary intervention. A cell-penetrating PAR1 pepducin, PZ-128, currently being tested as an antithrombotic agent in the acute setting in the TRIP-PCI study (Thrombin Receptor Inhibitory Pepducin-Percutaneous Coronary Intervention), caused a significant decrease in total atherosclerotic burden by 58% to 70% (P<0.05) and reduced plaque macrophage content by 54% (P<0.05) in apolipoprotein E-deficient mice. An MMP1 inhibitor gave similar beneficial effects, in contrast to the thrombin inhibitor bivalirudin that gave no improvement on atherosclerosis end points. Mechanistic studies revealed that inflammatory signaling mediated by MMP1-PAR1 plays a critical role in amplifying tumor necrosis factor α signaling in endothelial cells. CONCLUSIONS These data suggest that targeting the MMP1-PAR1 system may be effective in tamping down chronic inflammatory signaling in plaques and halting the progression of atherosclerosis.
Collapse
Affiliation(s)
- Rajashree Rana
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Tianfang Huang
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Georgios Koukos
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Elizabeth K Fletcher
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Susan E Turner
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Andrew Shearer
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Paul A Gurbel
- Inova Center for Thrombosis Research and Translational Medicine, Inova Heart and Vascular Institute, Inova Fairfax Hospital, Falls Church, VA (P.A.G., K.P.B.)
| | - Jeffrey J Rade
- Department of Medicine, Division of Cardiology, University of Massachusetts Memorial Medical Center, University of Massachusetts Medical School, Worcester (J.J.R.)
| | - Carey D Kimmelstiel
- Department of Medicine, Division of Cardiology, Tufts Medical Center, Boston, MA (C.D.K.)
| | - Kevin P Bliden
- Inova Center for Thrombosis Research and Translational Medicine, Inova Heart and Vascular Institute, Inova Fairfax Hospital, Falls Church, VA (P.A.G., K.P.B.)
| | - Lidija Covic
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Athan Kuliopulos
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| |
Collapse
|
47
|
Plumbagin Alleviates Capillarization of Hepatic Sinusoids In Vitro by Downregulating ET-1, VEGF, LN, and Type IV Collagen. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5603216. [PMID: 28770223 PMCID: PMC5523349 DOI: 10.1155/2017/5603216] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/28/2017] [Accepted: 06/05/2017] [Indexed: 12/19/2022]
Abstract
Critical roles for liver sinusoidal endothelial cells (LSECs) in liver fibrosis have been demonstrated, while little is known regarding the underlying molecular mechanisms of drugs delivered to the LSECs. Our previous study revealed that plumbagin plays an antifibrotic role in liver fibrosis. In this study, we investigated whether plumbagin alleviates capillarization of hepatic sinusoids by downregulating endothelin-1 (ET-1), vascular endothelial growth factor (VEGF), laminin (LN), and type IV collagen on leptin-stimulated LSECs. We found that normal LSECs had mostly open fenestrae and no organized basement membrane. Leptin-stimulated LSECs showed the formation of a continuous basement membrane with few open fenestrae, which were the features of capillarization. Expression of ET-1, VEGF, LN, and type IV collagen was enhanced in leptin-stimulated LSECs. Plumbagin was used to treat leptin-stimulated LSECs. The sizes and numbers of open fenestrae were markedly decreased, and no basement membrane production was found after plumbagin administration. Plumbagin decreased the levels of ET-1, VEGF, LN, and type IV collagen in leptin-stimulated LSECs. Plumbagin promoted downregulation of ET-1, VEGF, LN, and type IV collagen mRNA. Altogether, our data reveal that plumbagin reverses capillarization of hepatic sinusoids by downregulation of ET-1, VEGF, LN, and type IV collagen.
Collapse
|
48
|
An J, Zhang D, Wu J, Li J, Teng X, Gao X, Li R, Wang X, Xia L, Xia Y. The acitretin and methotrexate combination therapy for psoriasis vulgaris achieves higher effectiveness and less liver fibrosis. Pharmacol Res 2017; 121:158-168. [PMID: 28414177 DOI: 10.1016/j.phrs.2017.04.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/30/2017] [Accepted: 04/11/2017] [Indexed: 02/05/2023]
Abstract
Both acitretin and methotrexate are effective in ameliorating psoriatic lesion. However, their combination has been seldom reported in the treatment of psoriasis because of the warning regarding the potential hepatotoxicity of the drug interactions. This study was designed to investigate the effectiveness of such combination therapy for psoriasis vulgaris, and the potential benefit as well as side effect during the treatment. Thirty-nine patients with psoriasis vulgaris were treated with acitretin, methotrexate or their combination or as control. Similarly, K14-VEGF transgenic psoriasis-like mice were treated with these drugs. Human primary keratinocytes and hepatic stellate cells were used for analyzing their effect in vitro. The results showed that the combination therapy exhibited higher effectiveness in remitting skin lesion, but did not significantly affect the liver function of both patients and mice. Moreover, the combination groups showed less elevation of profibrotic factors in sera when compared with methotrexate alone groups accordingly. Furthermore, primary keratinocytes expressed more involucrin as well as loricrin and proliferated more slowly on the combined stimulation. Interestingly, such combination treatment induced lower expression of profibrotic factors in hepatic stellate cells. In conclusion, the acitretin-methotrexate combination therapy for psoriasis vulgaris can achieve higher effectiveness and result in less liver fibrosis.
Collapse
Affiliation(s)
- Jingang An
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Dingwei Zhang
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jiawen Wu
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jiong Li
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, China
| | - Xiu Teng
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, China
| | - Xiaomin Gao
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Ruilian Li
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Xiuying Wang
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Linlin Xia
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
49
|
Lin C, Borensztajn K, Spek CA. Targeting coagulation factor receptors - protease-activated receptors in idiopathic pulmonary fibrosis. J Thromb Haemost 2017; 15:597-607. [PMID: 28079978 DOI: 10.1111/jth.13623] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Indexed: 12/11/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal lung disease with a 5-year mortality rate of > 50% and unknown etiology. Treatment options remain limited and, currently, only two drugs are available, i.e. nintedanib and pirfenidone. However, both of these antifibrotic agents only slow down the progression of the disease, and do not remarkably prolong the survival of IPF patients. Hence, the discovery of new therapeutic targets for IPF is crucial. Studies exploring the mechanisms that are involved in IPF have identified several possible targets for therapeutic interventions. Among these, blood coagulation factor receptors, i.e. protease-activated receptors (PARs), are key candidates, as these receptors mediate the cellular effects of coagulation factors and play central roles in influencing inflammatory and fibrotic responses. In this review, we will focus on the controversial role of the coagulation cascade in the pathogenesis of IPF. In the light of novel data, we will attempt to reconciliate the apparently conflicting data and discuss the possibility of pharmacologic targeting of PARs for the treatment of fibroproliferative diseases.
Collapse
Affiliation(s)
- C Lin
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - K Borensztajn
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands
- Inserm UMR1152, Medical School Xavier Bichat, Paris, France
- Département Hospitalo-universtaire FIRE (Fibrosis, Inflammation and Remodeling) and LabEx Inflamex, Paris, France
| | - C A Spek
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| |
Collapse
|