1
|
Cline H, Wei Z, Groeneveld DJ, Hix JML, Xu X, Flick MJ, Palumbo JS, Poole LG, Dockendorff C, Griffin JH, Luyendyk JP. Hepatocyte-independent PAR1-biased signaling controls liver pathology in experimental obesity. J Thromb Haemost 2024; 22:3191-3198. [PMID: 39122189 PMCID: PMC11513232 DOI: 10.1016/j.jtha.2024.07.017] [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: 03/25/2024] [Revised: 07/05/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Protease-activated receptor-1 (PAR1) has emerged as an important link between coagulation and the complications of obesity including metabolic dysfunction-associated steatotic liver disease (MASLD). PAR1 is expressed by various cells and cleaved by different proteases to generate unique tethered agonists that activate distinct signaling pathways. Mice expressing PAR1 with an R41Q mutation have disabled canonical thrombin-mediated signaling, whereas R46Q mice express PAR1 resistant to noncanonical signaling by activated protein C. METHODS Mice with whole body and hepatocyte-selective PAR1 deficiency as well as PAR1 R41Q and R46Q mice were fed a high-fat diet (HFD) to induce MASLD. RESULTS HFD-fed R41Q mice displayed reduced hepatic steatosis and liver/body weight ratio. In contrast, HFD-fed R46Q mice displayed increased relative liver weight and hepatic steatosis alongside increased serum alanine aminotransferase activity. Surprisingly, despite the distinct impact of PAR1 mutations on steatosis, selective deletion of PAR1 in hepatocytes had no impact. To evaluate a viable PAR1-targeted approach, mice with HFD-induced obesity were treated with the allosteric PAR1 modulator NRD-21, which inhibits canonical PAR1 inflammatory signaling but promotes PAR1 protective, noncanonical anti-inflammatory signaling. NRD-21 treatment reduced plasma tumor necrosis factor-alpha, serum alanine aminotransferase activity, hepatic steatosis, and insulin resistance (Homeostatic Model Assessment for Insulin Resistance) but increased plasma active glucagon-like peptide-1. CONCLUSION The results suggest that nonhepatocellular canonical PAR1 cleavage drives MASLD in obese mice and provide translational proof-of-concept that selective pharmacologic modulation of PAR1 yields multiple metabolic benefits in experimental obesity.
Collapse
Affiliation(s)
- Holly Cline
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA
| | - Zimu Wei
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA
| | - Dafna J Groeneveld
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA
| | - Jeremy M L Hix
- Department of Radiology and Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, Michigan, USA
| | - Xiao Xu
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, California, USA
| | - Matthew J Flick
- Department of Pathology and Laboratory Medicine, University of North Carolina Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joseph S Palumbo
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Lauren G Poole
- Department of Pharmacology, Rutgers University, Piscataway, New Jersey, USA
| | | | - John H Griffin
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, California, USA
| | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA.
| |
Collapse
|
2
|
Wan X, Zhang W, Dai L, Chen L. The Role of Extracellular Vesicles in Bone Regeneration and Associated Bone Diseases. Curr Issues Mol Biol 2024; 46:9269-9285. [PMID: 39329900 PMCID: PMC11430372 DOI: 10.3390/cimb46090548] [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: 08/03/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024] Open
Abstract
Extracellular vesicles (EVs) are nanoscale particles with a lipid bilayer membrane structure secreted by various cell types. Nearly all human cells secrete EVs, primarily mediating intercellular communication. In recent years, scientists have discovered that EVs can carry multiple biological cargos, such as DNA, non-coding RNAs (ncRNAs), proteins, cytokines, and lipids, and mediate intercellular signal transduction. Bone is a connective tissue with a nerve supply and high vascularization. The repair process after injury is highly complex, involving interactions among multiple cell types and biological signaling pathways. Bone regeneration consists of a series of coordinated osteoconductive and osteoinductive biological processes. As mediators of intercellular communication, EVs can promote bone regeneration by regulating osteoblast-mediated bone formation, osteoclast-mediated bone resorption, and other pathways. This review summarizes the biogenesis of EVs and the mechanisms by which EV-mediated intercellular communication promotes bone regeneration. Additionally, we focus on the research progress of EVs in various diseases related to bone regeneration. Finally, based on the above research, we explore the clinical applications of engineered EVs in the diagnosis and treatment of bone regeneration-related diseases.
Collapse
Affiliation(s)
- Xinyue Wan
- School of Medicine, Chongqing University, Chongqing 400030, China; (X.W.); (W.Z.); (L.D.)
| | - Wenjie Zhang
- School of Medicine, Chongqing University, Chongqing 400030, China; (X.W.); (W.Z.); (L.D.)
| | - Lingyan Dai
- School of Medicine, Chongqing University, Chongqing 400030, China; (X.W.); (W.Z.); (L.D.)
| | - Liang Chen
- School of Medicine, Chongqing University, Chongqing 400030, China; (X.W.); (W.Z.); (L.D.)
- Department of Bone and Soft Tissue Oncology, Chongqing University Cancer Hospital, Chongqing University School of Medicine, Chongqing 400030, China
| |
Collapse
|
3
|
Buyl K, Vrints M, Fernando R, Desmae T, Van Eeckhoutte T, Jans M, Van Der Schueren J, Boeckmans J, Rodrigues RM, De Boe V, Rogiers V, De Kock J, Beirinckx F, Vanhaecke T. Human skin stem cell-derived hepatic cells as in vitro drug discovery model for insulin-driven de novo lipogenesis. Eur J Pharmacol 2023; 957:175989. [PMID: 37572939 DOI: 10.1016/j.ejphar.2023.175989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/14/2023]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), is characterized by intrahepatic triglyceride accumulation and can progress to metabolic dysfunction-associated steatohepatitis (MASH) and liver fibrosis. Hepatic de novo lipogenesis (DNL), activated by glucose and insulin, is a central pathway contributing to early-stage development of MASLD. The emerging global prevalence of MASLD highlights the urgent need for pharmaceutical intervention to combat this health threat. However, the identification of novel drugs that could inhibit hepatic DNL is hampered by a lack of reliable, insulin-sensitive, human, in vitro, hepatic models. Here, we report human skin stem cell-derived hepatic cells (hSKP-HPC) as a unique in vitro model to study insulin-driven DNL (iDNL), evidenced by both gene expression and lipid accumulation readouts. Insulin-sensitive hSKP-HPC showed increased sterol regulatory element-binding protein 1c (SREBP-1c) expression, a key transcription factor for DNL. Furthermore, this physiologically relevant in vitro human steatosis model allowed both inhibition and activation of the iDNL pathway using reference inhibitors and activators, respectively. Optimisation of the lipid accumulation assay to a high-throughput, 384-well format enabled the screening of a library of annotated compounds, delivering new insights on key players in the iDNL pathway and MASLD pathophysiology. Together, these results establish the value of the hSKP-HPC model in preclinical development of antisteatotic drugs to combat MASLD.
Collapse
Affiliation(s)
- Karolien Buyl
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium.
| | - Martine Vrints
- Galapagos NV, Industriepark Mechelen Noord, Generaal De Wittelaan L11 A3, B-2880, Mechelen, Belgium
| | - Ruani Fernando
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium
| | - Terry Desmae
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium
| | - Thomas Van Eeckhoutte
- Galapagos NV, Industriepark Mechelen Noord, Generaal De Wittelaan L11 A3, B-2880, Mechelen, Belgium
| | - Mia Jans
- Galapagos NV, Industriepark Mechelen Noord, Generaal De Wittelaan L11 A3, B-2880, Mechelen, Belgium
| | - Jan Van Der Schueren
- Galapagos NV, Industriepark Mechelen Noord, Generaal De Wittelaan L11 A3, B-2880, Mechelen, Belgium
| | - Joost Boeckmans
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium
| | - Robim M Rodrigues
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium
| | - Veerle De Boe
- Department of Urology, Universitair Ziekenhuis Brussel (UZ-Brussel), Laarbeeklaan 101, B-1090, Brussels, Belgium
| | - Vera Rogiers
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium
| | - Joery De Kock
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium
| | - Filip Beirinckx
- Galapagos NV, Industriepark Mechelen Noord, Generaal De Wittelaan L11 A3, B-2880, Mechelen, Belgium
| | - Tamara Vanhaecke
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium
| |
Collapse
|
4
|
Anton A, Shalaby S, Hernández-Gea V. Coagulation meets senescence in NASH! Hepatology 2023; 78:1023-1025. [PMID: 37246429 DOI: 10.1097/hep.0000000000000498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/30/2023]
Affiliation(s)
- Aina Anton
- Barcelona Hepatic Hemodynamic Laboratory, Liver Unit, Hospital Clínic Barcelona. Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Sarah Shalaby
- Barcelona Hepatic Hemodynamic Laboratory, Liver Unit, Hospital Clínic Barcelona. Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Virginia Hernández-Gea
- Barcelona Hepatic Hemodynamic Laboratory, Liver Unit, Hospital Clínic Barcelona. Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| |
Collapse
|
5
|
Hara T, Uemoto R, Sekine A, Mitsui Y, Masuda S, Yamagami H, Kurahashi K, Yoshida S, Otoda T, Yuasa T, Kuroda A, Ikeda Y, Endo I, Honda S, Yoshimoto K, Kondo A, Tamaki T, Matsumoto T, Matsuhisa M, Abe M, Aihara KI. Plasma Heparin Cofactor II Activity Is Inversely Associated with Hepatic Fibrosis of Non-Alcoholic Fatty Liver Disease in Patients with Type 2 Diabetes Mellitus. J Atheroscler Thromb 2023; 30:871-883. [PMID: 36244745 PMCID: PMC10406648 DOI: 10.5551/jat.63752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/12/2022] [Indexed: 08/04/2023] Open
Abstract
AIMS Thrombin exerts various pathophysiological functions by activating protease-activated receptors (PARs), and thrombin-induced activation of PARs promotes the development of non-alcoholic fatty liver disease (NAFLD). Since heparin cofactor II (HCII) specifically inactivates thrombin action, we hypothesized that plasma HCII activity correlates with the severity of NAFLD. METHODS A cross-sectional study was conducted. Plasma HCII activity and noninvasive clinical markers of hepatic fibrosis including fibrosis-4 (FIB-4) index, NAFLD fibrosis score (NFS) and aspartate aminotransferase-to-platelet ratio index (APRI) were determined in 305 Japanese patients with type 2 diabetes mellitus (T2DM). The relationships between plasma HCII activity and the clinical markers were statistically evaluated. RESULTS Multiple regression analysis including confounding factors showed that plasma HCII activity independently contributed to decreases in FIB-4 index (p<0.001), NFS (p<0.001) and APRI (p=0.004). In addition, logistic regression analysis for the prevalence of advanced hepatic fibrosis defined by the cutoff points of the clinical scores showed that plasma HCII activity was the sole and common negative factor for prevalence of advanced hepatic fibrosis (FIB-4 index: p=0.002, NFS: p=0.026 and APRI: p=0.012). CONCLUSIONS Plasma HCII activity was inversely associated with clinical hepatic fibrosis indices including FIB-4 index, NFS and APRI and with the prevalence of advanced hepatic fibrosis in patients with T2DM. The results suggest that HCII can serve as a novel biomarker for assessment of hepatic fibrosis of NAFLD in patients with T2DM.
Collapse
Affiliation(s)
- Tomoyo Hara
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Ryoko Uemoto
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Akiko Sekine
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yukari Mitsui
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Shiho Masuda
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Hiroki Yamagami
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kiyoe Kurahashi
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Sumiko Yoshida
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Toshiki Otoda
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Tomoyuki Yuasa
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Akio Kuroda
- Diabetes Therapeutics and Research Center, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Yasumasa Ikeda
- Department of Pharmacology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Itsuro Endo
- Department of Bioregulatory Sciences, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Soichi Honda
- Minami Municipal National Insurance Hospital, Tokushima, Japan
| | - Katsuhiko Yoshimoto
- Department of Medical Pharmacology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
- Kondo Naika Hospital, Tokushima, Japan
| | | | | | - Toshio Matsumoto
- Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Munehide Matsuhisa
- Diabetes Therapeutics and Research Center, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Masahiro Abe
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Ken-ichi Aihara
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
- Anan Medical Center, Tokushima, Japan
| |
Collapse
|
6
|
Airola C, Pallozzi M, Cerrito L, Santopaolo F, Stella L, Gasbarrini A, Ponziani FR. Microvascular Thrombosis and Liver Fibrosis Progression: Mechanisms and Clinical Applications. Cells 2023; 12:1712. [PMID: 37443746 PMCID: PMC10341358 DOI: 10.3390/cells12131712] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Fibrosis is an unavoidable consequence of chronic inflammation. Extracellular matrix deposition by fibroblasts, stimulated by multiple pathways, is the first step in the onset of chronic liver disease, and its propagation promotes liver dysfunction. At the same time, chronic liver disease is characterized by alterations in primary and secondary hemostasis but unlike previously thought, these changes are not associated with an increased risk of bleeding complications. In recent years, the role of coagulation imbalance has been postulated as one of the main mechanisms promoting hepatic fibrogenesis. In this review, we aim to investigate the function of microvascular thrombosis in the progression of liver disease and highlight the molecular and cellular networks linking hemostasis to fibrosis in this context. We analyze the predictive and prognostic role of coagulation products as biomarkers of liver decompensation (ascites, variceal hemorrhage, and hepatic encephalopathy) and liver-related mortality. Finally, we evaluate the current evidence on the application of antiplatelet and anticoagulant therapies for prophylaxis of hepatic decompensation or prevention of the progression of liver fibrosis.
Collapse
Affiliation(s)
- Carlo Airola
- Hepatology Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Maria Pallozzi
- Hepatology Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Lucia Cerrito
- Hepatology Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Francesco Santopaolo
- Hepatology Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Leonardo Stella
- Hepatology Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Antonio Gasbarrini
- Hepatology Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (F.S.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Hepatology Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (F.S.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
7
|
Hara T, Sata M, Fukuda D. Emerging roles of protease-activated receptors in cardiometabolic disorders. J Cardiol 2023; 81:337-346. [PMID: 36195252 DOI: 10.1016/j.jjcc.2022.09.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/05/2022]
Abstract
Cardiometabolic disorders, including obesity-related insulin resistance and atherosclerosis, share sterile chronic inflammation as a major cause; however, the precise underlying mechanisms of chronic inflammation in cardiometabolic disorders are not fully understood. Accumulating evidence suggests that several coagulation proteases, including thrombin and activated factor X (FXa), play an important role not only in the coagulation cascade but also in the proinflammatory responses through protease-activated receptors (PARs) in many cell types. Four members of the PAR family have been cloned (PAR 1-4). For instance, thrombin activates PAR-1, PAR-3, and PAR-4. FXa activates both PAR-1 and PAR-2, while it has no effect on PAR-3 or PAR-4. Previous studies demonstrated that PAR-1 and PAR-2 activated by thrombin or FXa promote gene expression of inflammatory molecules mainly via the NF-κB and ERK1/2 pathways. In obese adipose tissue and atherosclerotic vascular tissue, various stresses increase the expression of tissue factor and procoagulant activity. Recent studies indicated that the activation of PARs in adipocytes and vascular cells by coagulation proteases promotes inflammation in these tissues, which leads to the development of cardiometabolic diseases. This review briefly summarizes the role of PARs and coagulation proteases in the pathogenesis of inflammatory diseases and describes recent findings (including ours) on the potential participation of this system in the development of cardiometabolic disorders. New insights into PARs may ensure a better understanding of cardiometabolic disorders and suggest new therapeutic options for these major health threats.
Collapse
Affiliation(s)
- Tomoya Hara
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Daiju Fukuda
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan; Department of Cardiovascular Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
8
|
Kostoff RN, Briggs MB, Kanduc D, Dewanjee S, Kandimalla R, Shoenfeld Y, Porter AL, Tsatsakis A. Modifiable contributing factors to COVID-19: A comprehensive review. Food Chem Toxicol 2023; 171:113511. [PMID: 36450305 PMCID: PMC9701571 DOI: 10.1016/j.fct.2022.113511] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/29/2022]
Abstract
The devastating complications of coronavirus disease 2019 (COVID-19) result from an individual's dysfunctional immune response following the initial severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Multiple toxic stressors and behaviors contribute to underlying immune system dysfunction. SARS-CoV-2 exploits the dysfunctional immune system to trigger a chain of events ultimately leading to COVID-19. The current study identifies eighty immune system dysfunction-enabling toxic stressors and behaviors (hereafter called modifiable contributing factors (CFs)) that also link directly to COVID-19. Each CF is assigned to one of the five categories in the CF taxonomy shown in Section 3.3.: Lifestyle (e.g., diet, substance abuse); Iatrogenic (e.g., drugs, surgery); Biotoxins (e.g., micro-organisms, mycotoxins); Occupational/Environmental (e.g., heavy metals, pesticides); Psychosocial/Socioeconomic (e.g., chronic stress, lower education). The current study shows how each modifiable factor contributes to decreased immune system capability, increased inflammation and coagulation, and increased neural damage and neurodegeneration. It is unclear how real progress can be made in combatting COVID-19 and other similar diseases caused by viral variants without addressing and eliminating these modifiable CFs.
Collapse
Affiliation(s)
- Ronald Neil Kostoff
- Independent Consultant, Gainesville, VA, 20155, USA,Corresponding author. Independent Consultant, 13500 Tallyrand Way, Gainesville, VA, 20155, USA
| | | | - Darja Kanduc
- Dept. of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Via Orabona 4, Bari, 70125, Italy
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Ramesh Kandimalla
- Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, 500007, Telangana, India
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, 5265601, Israel
| | - Alan L. Porter
- School of Public Policy, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003, Heraklion, Greece
| |
Collapse
|
9
|
Hur WS, King KC, Patel YN, Nguyen YV, Wei Z, Yang Y, Juang LJ, Leung J, Kastrup CJ, Wolberg AS, Luyendyk JP, Flick MJ. Elimination of fibrin polymer formation or crosslinking, but not fibrinogen deficiency, is protective against diet-induced obesity and associated pathologies. J Thromb Haemost 2022; 20:2873-2886. [PMID: 36111375 PMCID: PMC9669152 DOI: 10.1111/jth.15877] [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: 07/27/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND Obesity predisposes individuals to metabolic syndrome, which increases the risk of cardiovascular diseases, non-alcoholic fatty liver disease (NAFLD), and type 2 diabetes. A pathological manifestation of obesity is the activation of the coagulation system. In turn, extravascular fibrin(ogen) deposits accumulate in adipose tissues and liver. These deposits promote adiposity and downstream sequelae by driving pro-inflammatory macrophage function through binding the leukocyte integrin receptor αM β2 . OBJECTIVES An unresolved question is whether conversion of soluble fibrinogen to a crosslinked fibrin matrix is required to exacerbate obesity-driven diseases. METHODS Here, fibrinogen-deficient/depleted mice (Fib- or treated with siRNA against fibrinogen [siFga]), mice expressing fibrinogen that cannot polymerize to fibrin (FibAEK ), and mice deficient in the fibrin crosslinking transglutaminase factor XIII (FXIII-) were challenged with a high-fat diet (HFD) and compared to mice expressing a mutant form of fibrinogen lacking the αM β2 -binding domain (Fib𝛾390-396A ). RESULTS AND CONCLUSIONS Consistent with prior studies, Fib𝛾390-396A mice were significantly protected from increased adiposity, NAFLD, hypercholesterolemia, and diabetes while Fib- and siFga-treated mice gained as much weight and developed obesity-associated pathologies identical to wildtype mice. FibAEK and FXIII- mice displayed an intermediate phenotype with partial protection from some obesity-associated pathologies. Results here indicate that fibrin(ogen) lacking αM β2 binding function offers substantial protection from obesity and associated disease that is partially recapitulated by preventing fibrin polymer formation or crosslinking of the wildtype molecule, but not by reduction or complete elimination of fibrinogen. Finally, these findings support the concept that fibrin polymerization and crosslinking are required for the full implementation of fibrin-driven inflammation in obesity.
Collapse
Affiliation(s)
- Woosuk S. Hur
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katharine C. King
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yesha N. Patel
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Y-Van Nguyen
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zimu Wei
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - Yi Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lih Jiin Juang
- Michael Smith Laboratories, and Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Jerry Leung
- Michael Smith Laboratories, and Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Christian J. Kastrup
- Michael Smith Laboratories, and Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
- Blood Research institute, Versiti, Milwaukee, WI, USA
| | - Alisa S Wolberg
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - James P Luyendyk
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - Matthew J. Flick
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
10
|
Ogresta D, Mrzljak A, Cigrovski Berkovic M, Bilic-Curcic I, Stojsavljevic-Shapeski S, Virovic-Jukic L. Coagulation and Endothelial Dysfunction Associated with NAFLD: Current Status and Therapeutic Implications. J Clin Transl Hepatol 2022; 10:339-355. [PMID: 35528987 PMCID: PMC9039716 DOI: 10.14218/jcth.2021.00268] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/24/2021] [Accepted: 10/08/2021] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is closely related to insulin resistance, type 2 diabetes mellitus and obesity. It is considered a multisystem disease and there is a strong association with cardiovascular disease and arterial hypertension, which interfere with changes in the coagulation system. Coagulation disorders are common in patients with hepatic impairment and are dependent on the degree of liver damage. Through a review of the literature, we consider and discuss possible disorders in the coagulation cascade and fibrinolysis, endothelial dysfunction and platelet abnormalities in patients with NAFLD.
Collapse
Affiliation(s)
- Doris Ogresta
- Department of Gastroenterology and Hepatology, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Anna Mrzljak
- Department of Gastroenterology and Hepatology, University Hospital Center Zagreb, Zagreb, Croatia
- Department of Medicine, University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Maja Cigrovski Berkovic
- Department for Endocrinology, Diabetes and Pharmacology, University Hospital Dubrava, Zagreb, Croatia
- Department of Kinesiological Anthropology and Methodology, Faculty of Kinesiology, University of Zagreb
- Department of Pharmacology, Faculty of Medicine, University of JJ Strossmayer, Osijek, Croatia
| | - Ines Bilic-Curcic
- Department of Pharmacology, Faculty of Medicine, University of JJ Strossmayer, Osijek, Croatia
- Department of Diabetes, Endocrinology and Metabolism Disorders, University Hospital Osijek, Osijek, Croatia
| | | | - Lucija Virovic-Jukic
- Department of Gastroenterology and Hepatology, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
- Department of Medicine, University of Zagreb, School of Medicine, Zagreb, Croatia
| |
Collapse
|
11
|
Wu F, Zhuang P, Zhang Y, Zhan C, Zhang Y, Jiao J. Egg and Dietary Cholesterol Consumption and Mortality Among Hypertensive Patients: Results From a Population-Based Nationwide Study. Front Nutr 2021; 8:739533. [PMID: 34778336 PMCID: PMC8588794 DOI: 10.3389/fnut.2021.739533] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 09/29/2021] [Indexed: 01/25/2023] Open
Abstract
Background: Hypertensive patients are sensitive to the amount of dietary cholesterol intake, especially cholesterol from the whole eggs. Whether whole egg and dietary cholesterol consumption are suitable for hypertensive patients is still controversial. Aim: The objective of the study was to examine the associations of intake of eggs as well as the dietary cholesterol with total mortality in a Chinese nationwide cohort. Methods: We utilized data from the China Health and Nutrition Survey (CHNS) from the year of 1991 to 2015. Cumulative averages of egg and cholesterol intake were calculated to represent the consumption of the long-term diet of the participants in each available round of the survey. Cox regression models were employed to estimate the effects of eggs and dietary cholesterol from the different sources on mortality among hypertensive patients. Results: A total of 8,095 participants were included in the final analysis and followed up for a mean of 11.4 years. Finally, 927 cases of death were detected. After adjustment for the multivariate factors, consuming more than seven eggs per week was related to 29% lower mortality among the hypertensive patients compared with the consumers with not more than two eggs per week [hazard ratio (HR): 0.71; 95% CI: 0.59–0.85; P < 0.001]. Similarly, the egg-sourced cholesterol intake was inversely associated with mortality (P = 0.002) whereas intake of the dietary cholesterol from the non-egg sources was significantly related to the higher mortality (P < 0.001). However, total cholesterol intake was not related to mortality among hypertensive patients. Substituting eggs for an equivalent amount of non-egg-sourced protein-abundant foods was also associated with lower mortality. Conclusion: Higher consumption of eggs and egg-sourced dietary cholesterol was associated with lower mortality among the enrolled Chinese hypertensive patients but non-egg-sourced cholesterol intake was related to higher mortality. Therefore, our findings do not support the view that hypertensive patients should avoid whole egg consumption for the purpose of restricting dietary cholesterol intake.
Collapse
Affiliation(s)
- Fei Wu
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pan Zhuang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - Yiju Zhang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - Chuchu Zhan
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - Yu Zhang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - Jingjing Jiao
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
12
|
Till Death Do Us Part-The Multifaceted Role of Platelets in Liver Diseases. Int J Mol Sci 2021; 22:ijms22063113. [PMID: 33803718 PMCID: PMC8003150 DOI: 10.3390/ijms22063113] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/09/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
Platelets are tightly connected with the liver, as both their production and their clearance are mediated by the liver. Platelets, in return, participate in a variety of liver diseases, ranging from non-alcoholic fatty liver diseases, (viral) hepatitis, liver fibrosis and hepatocellular carcinoma to liver regeneration. Due to their versatile functions, which include (1) regulation of hemostasis, (2) fine-tuning of immune responses and (3) release of growth factors and cellular mediators, platelets quickly adapt to environmental changes and modulate disease development, leading to different layers of complexity. Depending on the (patho)physiological context, platelets exert both beneficial and detrimental functions. Understanding the precise mechanisms through which platelet function is regulated at different stages of liver diseases and how platelets interact with various resident and non-resident liver cells helps to draw a clear picture of platelet-related therapeutic interventions. Therefore, this review summarizes the current knowledge on platelets in acute and chronic liver diseases and aims to shed light on how the smallest cells in the circulatory system account for changes in the (patho)physiology of the second largest organ in the human body.
Collapse
|
13
|
Noguchi D, Kuriyama N, Hibi T, Maeda K, Shinkai T, Gyoten K, Hayasaki A, Fujii T, Iizawa Y, Tanemura A, Murata Y, Kishiwada M, Sakurai H, Mizuno S. The Impact of Dabigatran Treatment on Sinusoidal Protection Against Hepatic Ischemia/Reperfusion Injury in Mice. Liver Transpl 2021; 27:363-384. [PMID: 33108682 PMCID: PMC7984054 DOI: 10.1002/lt.25929] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/30/2020] [Accepted: 10/17/2020] [Indexed: 12/27/2022]
Abstract
Thrombin is a key player in the coagulation cascade, and it is attracting much attention as a promotor of cellular injured signaling. In ischemia/reperfusion injury (IRI), which is a severe complication of liver transplantation, thrombin may also promote tissue damage. The aim of this study is to reveal whether dabigatran, a direct thrombin inhibitor, can attenuate hepatic IRI with focusing on a protection of sinusoidal endothelial cells (SECs). Both clinical patients who underwent hepatectomy and in vivo mice model of 60-minute hepatic partial-warm IRII, thrombin generation was evaluated before and after IRI. In next study, IRI mice were treated with or without dabigatran. In addition, hepatic SECs and hepatocytes pretreated with or without dabigatran were incubated in hypoxia/reoxygenation (H-R) environment in vitro. Thrombin generation evaluated by thrombin-antithrombin complex (TAT) was significantly enhanced after IRI in the clinical study and in vivo study. Thrombin exacerbated lactate dehydrogenase cytotoxicity levels in a dose-dependent manner in vitro. In an IRI model of mice, dabigatran treatment significantly improved liver histological damage, induced sinusoidal protection, and provided both antiapoptotic and anti-inflammatory effects. Furthermore, dabigatran not only enhanced endogenous thrombomodulin (TM) but also reduced excessive serum high-mobility group box-1 (HMGB-1). In H-R models of SECs, not hepatocytes, pretreatment with dabigatran markedly attenuated H-R damage, enhanced TM expression in cell lysate, and decreased extracellular HMGB-1. The supernatant of SECs pretreated with dabigatran protected hepatocytes from H-R damage and cellular death. Thrombin exacerbated hepatic IRI, and excessive extracellular HMGB-1 caused severe inflammation-induced and apoptosis-induced liver damage. In this situation, dabigatran treatment improved vascular integrity via sinusoidal protection and degraded HMGB-1 by endogenous TM enhancement on SECs, greatly ameliorating hepatic IRI.
Collapse
Affiliation(s)
- Daisuke Noguchi
- Department of Hepatobiliary Pancreatic and Transplant SurgeryMie University Graduate School of MedicineTsu cityMieJapan
| | - Naohisa Kuriyama
- Department of Hepatobiliary Pancreatic and Transplant SurgeryMie University Graduate School of MedicineTsu cityMieJapan
| | - Taemi Hibi
- Department of Hepatobiliary Pancreatic and Transplant SurgeryMie University Graduate School of MedicineTsu cityMieJapan
| | - Koki Maeda
- Department of Hepatobiliary Pancreatic and Transplant SurgeryMie University Graduate School of MedicineTsu cityMieJapan
| | - Toru Shinkai
- Department of Hepatobiliary Pancreatic and Transplant SurgeryMie University Graduate School of MedicineTsu cityMieJapan
| | - Kazuyuki Gyoten
- Department of Hepatobiliary Pancreatic and Transplant SurgeryMie University Graduate School of MedicineTsu cityMieJapan
| | - Aoi Hayasaki
- Department of Hepatobiliary Pancreatic and Transplant SurgeryMie University Graduate School of MedicineTsu cityMieJapan
| | - Takehiro Fujii
- Department of Hepatobiliary Pancreatic and Transplant SurgeryMie University Graduate School of MedicineTsu cityMieJapan
| | - Yusuke Iizawa
- Department of Hepatobiliary Pancreatic and Transplant SurgeryMie University Graduate School of MedicineTsu cityMieJapan
| | - Akihiro Tanemura
- Department of Hepatobiliary Pancreatic and Transplant SurgeryMie University Graduate School of MedicineTsu cityMieJapan
| | - Yasuhiro Murata
- Department of Hepatobiliary Pancreatic and Transplant SurgeryMie University Graduate School of MedicineTsu cityMieJapan
| | - Masashi Kishiwada
- Department of Hepatobiliary Pancreatic and Transplant SurgeryMie University Graduate School of MedicineTsu cityMieJapan
| | - Hiroyuki Sakurai
- Department of Hepatobiliary Pancreatic and Transplant SurgeryMie University Graduate School of MedicineTsu cityMieJapan
| | - Shugo Mizuno
- Department of Hepatobiliary Pancreatic and Transplant SurgeryMie University Graduate School of MedicineTsu cityMieJapan
| |
Collapse
|
14
|
Deng YX, He WG, Cai HJ, Jiang JH, Yang YY, Dan YR, Luo HH, Du Y, Chen L, He BC. Analysis and Validation of Hub Genes in Blood Monocytes of Postmenopausal Osteoporosis Patients. Front Endocrinol (Lausanne) 2021; 12:815245. [PMID: 35095774 PMCID: PMC8792966 DOI: 10.3389/fendo.2021.815245] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/13/2021] [Indexed: 01/02/2023] Open
Abstract
Osteoporosis is a common systemic bone disease caused by the imbalance between osteogenic activity and osteoclastic activity. Aged women are at higher risk of osteoporosis, partly because of estrogen deficiency. However, the underlying mechanism of how estrogen deficiency affects osteoclast activity has not yet been well elucidated. In this study, GSE2208 and GSE56815 datasets were downloaded from GEO database with 25 PreH BMD women and 25 PostL BMD women in total. The RRA algorithm determined 38 downregulated DEGs and 30 upregulated DEGs. Through GO analysis, we found that downregulated DEGs were mainly enriched in myeloid cell differentiation, cytokine-related functions while upregulated DEGs enriched in immune-related biological processes; pathways like Notch signaling and MAPK activation were found in KEGG/Rectome pathway database; a PPI network which contains 66 nodes and 91 edges was constructed and three Modules were obtained by Mcode; Correlation analysis helped us to find highly correlated genes in each module. Moreover, three hub genes FOS, PTPN6, and CTSD were captured by Cytohubba. Finally, the hub genes were further confirmed in blood monocytes of ovariectomy (OVX) rats by real-time PCR assay. In conclusion, the integrative bioinformatics analysis and real-time PCR analysis were utilized to offer fresh light into the role of monocytes in premenopausal osteoporosis and identified FOS, PTPN6, and CTSD as potential biomarkers for postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Yi-Xuan Deng
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
| | - Wen-Ge He
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
- Department of Orthopaedics, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Bone and Soft Tissue Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Hai-Jun Cai
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
| | - Jin-Hai Jiang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
| | - Yuan-Yuan Yang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
| | - Yan-Rong Dan
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
| | - Hong-Hong Luo
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
| | - Yu Du
- Department of Orthopaedics, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Liang Chen
- Department of Orthopaedics, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Bone and Soft Tissue Oncology, Chongqing University Cancer Hospital, Chongqing, China
- *Correspondence: Liang Chen, ; Bai-Cheng He,
| | - Bai-Cheng He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
- *Correspondence: Liang Chen, ; Bai-Cheng He,
| |
Collapse
|
15
|
Zhang L, She ZG, Li H, Zhang XJ. Non-alcoholic fatty liver disease: a metabolic burden promoting atherosclerosis. Clin Sci (Lond) 2020; 134:1775-1799. [PMID: 32677680 DOI: 10.1042/cs20200446] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/06/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the fastest growing chronic liver disease, with a prevalence of up to 25% worldwide. Individuals with NAFLD have a high risk of disease progression to cirrhosis, hepatocellular carcinoma (HCC), and liver failure. With the exception of intrahepatic burden, cardiovascular disease (CVD) and especially atherosclerosis (AS) are common complications of NAFLD. Furthermore, CVD is a major cause of death in NAFLD patients. Additionally, AS is a metabolic disorder highly associated with NAFLD, and individual NAFLD pathologies can greatly increase the risk of AS. It is increasingly clear that AS-associated endothelial cell damage, inflammatory cell activation, and smooth muscle cell proliferation are extensively impacted by NAFLD-induced systematic dyslipidemia, inflammation, oxidative stress, the production of hepatokines, and coagulations. In clinical trials, drug candidates for NAFLD management have displayed promising effects for the treatment of AS. In this review, we summarize the key molecular events and cellular factors contributing to the metabolic burden induced by NAFLD on AS, and discuss therapeutic strategies for the improvement of AS in individuals with NAFLD.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Luojia Mount Wuchang, Wuhan 430072, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Luojia Mount Wuchang, Wuhan 430072, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Luojia Mount Wuchang, Wuhan 430072, China
- Basic Medical School, Wuhan University, Wuhan 430071, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Luojia Mount Wuchang, Wuhan 430072, China
| |
Collapse
|
16
|
Poole LG, Pant A, Cline‐Fedewa HM, Williams KJ, Copple BL, Palumbo JS, Luyendyk JP. Liver fibrosis is driven by protease-activated receptor-1 expressed by hepatic stellate cells in experimental chronic liver injury. Res Pract Thromb Haemost 2020; 4:906-917. [PMID: 32685902 PMCID: PMC7354391 DOI: 10.1002/rth2.12403] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 05/14/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Blood coagulation protease activity is proposed to drive hepatic fibrosis through activation of protease-activated receptors (PARs). Whole-body PAR-1 deficiency reduces experimental hepatic fibrosis, and in vitro studies suggest a potential contribution by PAR-1 expressed by hepatic stellate cells. However, owing to a lack of specific tools, the cell-specific role of PAR-1 in experimental hepatic fibrosis has never been formally investigated. Using a novel mouse expressing a conditional PAR-1 allele, we tested the hypothesis that PAR-1 expressed by hepatic stellate cells contributes to hepatic fibrosis. METHODS PAR-1flox/flox mice were crossed with mice expressing Cre recombinase controlled by the lecithin retinol acyltransferase (LRAT) promoter, which induces recombination in hepatic stellate cells. Male PAR-1flox/flox/LRATCre and PAR-1flox/flox mice were challenged twice weekly with carbon tetrachloride (CCl4, 1 mL/kg i.p.) for 6 weeks to induce liver fibrosis. RESULTS PAR-1 mRNA levels were reduced (>95%) in hepatic stellate cells isolated from PAR-1flox/flox/LRATCre mice. Hepatic stellate cell activation was evident in CCl4-challenged PAR-1flox/flox mice, indicated by increased α-smooth muscle actin labeling and induction of several profibrogenic genes. CCl4-challenged PAR-1flox/flox mice displayed robust hepatic collagen deposition, indicated by picrosirius red staining and type I collagen immunolabeling. Notably, stellate cell activation and collagen deposition were significantly reduced (>30%) in PAR-1flox/flox/LRATCre mice. Importantly, the reduction in liver fibrosis was not a consequence of reduced acute CCl4 hepatotoxicity in PAR-1flox/flox/LRATCre mice. CONCLUSIONS The results constitute the first direct experimental evidence that PAR-1 expressed by stellate cells directly promotes their profibrogenic phenotype and hepatic fibrosis in vivo.
Collapse
Affiliation(s)
- Lauren G. Poole
- Institute for Integrative ToxicologyMichigan State UniversityEast LansingMIUSA
- Department of Pathobiology & Diagnostic InvestigationMichigan State UniversityEast LansingMIUSA
| | - Asmita Pant
- Department of Pathobiology & Diagnostic InvestigationMichigan State UniversityEast LansingMIUSA
| | - Holly M. Cline‐Fedewa
- Department of Pathobiology & Diagnostic InvestigationMichigan State UniversityEast LansingMIUSA
| | - Kurt J. Williams
- Department of Pathobiology & Diagnostic InvestigationMichigan State UniversityEast LansingMIUSA
| | - Bryan L. Copple
- Department of Pharmacology and ToxicologyMichigan State UniversityEast LansingMIUSA
| | - Joseph S. Palumbo
- Cancer and Blood Diseases InstituteCincinnati Children’s Hospital Medical Center and the University of Cincinnati College of MedicineCincinnatiOHUSA
| | - James P. Luyendyk
- Institute for Integrative ToxicologyMichigan State UniversityEast LansingMIUSA
- Department of Pathobiology & Diagnostic InvestigationMichigan State UniversityEast LansingMIUSA
- Department of Pharmacology and ToxicologyMichigan State UniversityEast LansingMIUSA
| |
Collapse
|
17
|
Decreased M1 macrophage polarization in dabigatran-treated Ldlr-deficient mice: Implications for atherosclerosis and adipose tissue inflammation. Atherosclerosis 2019; 287:81-88. [DOI: 10.1016/j.atherosclerosis.2019.06.897] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 05/14/2019] [Accepted: 06/07/2019] [Indexed: 12/22/2022]
|
18
|
Luyendyk JP, Schoenecker JG, Flick MJ. The multifaceted role of fibrinogen in tissue injury and inflammation. Blood 2019; 133:511-520. [PMID: 30523120 PMCID: PMC6367649 DOI: 10.1182/blood-2018-07-818211] [Citation(s) in RCA: 336] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 11/26/2018] [Indexed: 02/08/2023] Open
Abstract
The canonical role of the hemostatic and fibrinolytic systems is to maintain vascular integrity. Perturbations in either system can prompt primary pathological end points of hemorrhage or thrombosis with vessel occlusion. However, fibrin(ogen) and proteases controlling its deposition and clearance, including (pro)thrombin and plasmin(ogen), have powerful roles in driving acute and reparative inflammatory pathways that affect the spectrum of tissue injury, remodeling, and repair. Indeed, fibrin(ogen) deposits are a near-universal feature of tissue injury, regardless of the nature of the inciting event, including injuries driven by mechanical insult, infection, or immunological derangements. Fibrin can modify multiple aspects of inflammatory cell function by engaging leukocytes through a variety of cellular receptors and mechanisms. Studies on the role of coagulation system activation and fibrin(ogen) deposition in models of inflammatory disease and tissue injury have revealed points of commonality, as well as context-dependent contributions of coagulation and fibrinolytic factors. However, there remains a critical need to define the precise temporal and spatial mechanisms by which fibrinogen-directed inflammatory events may dictate the severity of tissue injury and coordinate the remodeling and repair events essential to restore normal organ function. Current research trends suggest that future studies will give way to the identification of novel hemostatic factor-targeted therapies for a range of tissue injuries and disease.
Collapse
Affiliation(s)
- James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation
- Department of Pharmacology and Toxicology, and
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI
| | - Jonathan G Schoenecker
- Department of Orthopaedics
- Department of Pharmacology
- Department of Pediatrics, and
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN; and
| | - Matthew J Flick
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
19
|
Posma JJ, Grover SP, Hisada Y, Owens AP, Antoniak S, Spronk HM, Mackman N. Roles of Coagulation Proteases and PARs (Protease-Activated Receptors) in Mouse Models of Inflammatory Diseases. Arterioscler Thromb Vasc Biol 2019; 39:13-24. [PMID: 30580574 PMCID: PMC6310042 DOI: 10.1161/atvbaha.118.311655] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/19/2018] [Indexed: 12/20/2022]
Abstract
Activation of the blood coagulation cascade leads to fibrin deposition and platelet activation that are required for hemostasis. However, aberrant activation of coagulation can lead to thrombosis. Thrombi can cause tissue ischemia, and fibrin degradation products and activated platelets can enhance inflammation. In addition, coagulation proteases activate cells by cleavage of PARs (protease-activated receptors), including PAR1 and PAR2. Direct oral anticoagulants have recently been developed to specifically inhibit the coagulation proteases FXa (factor Xa) and thrombin. Administration of these inhibitors to wild-type mice can be used to determine the roles of FXa and thrombin in different inflammatory diseases. These results can be compared with the phenotypes of mice with deficiencies of either Par1 (F2r) or Par2 (F2rl1). However, inhibition of coagulation proteases will have effects beyond reducing PAR signaling, and a deficiency of PARs will abolish signaling from all proteases that activate these receptors. We will summarize studies that examine the roles of coagulation proteases, particularly FXa and thrombin, and PARs in different mouse models of inflammatory disease. Targeting FXa and thrombin or PARs may reduce inflammatory diseases in humans.
Collapse
Affiliation(s)
- Jens J Posma
- Laboratory for Clinical Thrombosis and Hemostasis, Departments of Internal Medicine and Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Steven P Grover
- Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yohei Hisada
- Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - A. Phillip Owens
- Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, OH, USA
| | - Silvio Antoniak
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Henri M Spronk
- Laboratory for Clinical Thrombosis and Hemostasis, Departments of Internal Medicine and Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Nigel Mackman
- Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
20
|
Bulani Y, Sharma SS. Argatroban Attenuates Diabetic Cardiomyopathy in Rats by Reducing Fibrosis, Inflammation, Apoptosis, and Protease-Activated Receptor Expression. Cardiovasc Drugs Ther 2018; 31:255-267. [PMID: 28695302 DOI: 10.1007/s10557-017-6732-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE Chronic diabetes is associated with cardiovascular dysfunctions. Diabetic cardiomyopathy (DCM) is one of the serious cardiovascular complications associated with diabetes. Despite significant efforts in understanding the pathophysiology of DCM, management of DCM is not adequate due to its complex pathophysiology. Recently, involvement of protease-activated receptors (PARs) has been postulated in cardiovascular diseases. These receptors are activated by thrombin, trypsin, or other serine proteases. Expression of PAR has been shown to be increased in cardiac diseases such as myocardial infarction, viral myocarditis, and pulmonary arterial hypertension. However, the role of PAR in DCM has not been elucidated yet. Therefore, in the present study, we have investigated the role of PAR in the condition of DCM using a pharmacological approach. We used argatroban, a direct thrombin inhibitor for targeting PAR. METHODS Type-2 diabetes mellitus (T2DM) was induced by high-fat feeding along with low dose streptozotocin (STZ 35 mg/kg, i.p. single dose) in male Sprague-Dawley rats. After 16 weeks of diabetes induction, animals were treated with argatroban at 0.3 and 1 mg/kg dose daily for 4 weeks. After 20 weeks, ventricular functions were measured using ventricular catheterization. Cardiac histology, TUNEL staining, and immunoblotting were performed to evaluate cardiac fibrosis, DNA fragmentation, and expression level of different proteins, respectively. RESULTS T2DM was associated with cardiac structural and functional disturbances as evidenced from impaired cardiac functional parameters and increased fibrosis. There was a significant increase in PAR expression after 20 weeks of diabetes induction. Four weeks argatroban treatment ameliorated metabolic alterations (reduced plasma glucose and cholesterol), ventricular dysfunctions (improved systolic and diastolic functions), cardiac fibrosis (reduced percentage area of collagen in picro-sirius red staining), and apoptosis (reduced TUNEL positive nuclei). Reduced expression of PAR1 and PAR4 in the argatroban-treated group indicates a response towards inhibition of thrombin. In addition, AKT (Ser-473), GSK-3β (Ser-9), p-65 NFĸB phosphorylation, TGF-β, COX-2, and caspase-3 expression were reduced significantly along with an increase in SERCA expression in argatroban-treated diabetic rats which indicated the anti-fibrotic, anti-inflammatory, and anti-apoptotic potential of argatroban in DCM. CONCLUSION This study suggests the ameliorative effects of argatroban in diabetic cardiomyopathy by improving ventricular functions and reducing fibrosis, inflammation, apoptosis, and PAR expression.
Collapse
Affiliation(s)
- Yogesh Bulani
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector-67, SAS, Nagar (Mohali), Punjab, 160062, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector-67, SAS, Nagar (Mohali), Punjab, 160062, India.
| |
Collapse
|
21
|
Bulani Y, Srinivasan K, Sharma SS. Attenuation of type-1 diabetes-induced cardiovascular dysfunctions by direct thrombin inhibitor in rats: a mechanistic study. Mol Cell Biochem 2018; 451:69-78. [PMID: 29971544 DOI: 10.1007/s11010-018-3394-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 06/26/2018] [Indexed: 01/04/2023]
Abstract
Chronic diabetes is associated with ventricular dysfunctions in the absence of hypertension and coronary artery diseases. This condition is termed as diabetic cardiomyopathy (DCM). There is no favourable treatment available for the management of diabetic cardiomyopathy. Recent studies have reported increase in circulating thrombin level among diabetic patients which is responsible for hypercoagulability of blood. Thrombin induces inflammation and fibrosis, and enhances cardiac cell growth and contractility in vitro. In this study, we have investigated the effects of argatroban; a direct thrombin inhibitor against DCM in streptozotocin-induced type-1 diabetes. Diabetes was induced by single dose of streptozotocin (STZ; 50 mg/kg, i.p.) in male Sprague-Dawley rats. After 4 weeks of diabetes induction, the animals were treated with argatroban (0.3 and 1 mg/kg, i.p. daily) for the next 4 weeks. The effect of argatroban was evaluated against diabetes-associated cardiac dysfunction, structural alteration and protein expression. STZ-induced diabetic rats exhibited significant decline in left ventricular functions. Four weeks of treatments with argatroban significantly improved ventricular functions without affecting heart rate. Further, it also protected heart against structural changes induced by diabetes as shown by reduction in fibrosis, hypertrophy and apoptosis. The improvement in cardiac functions and structural changes was associated with significant reduction in left ventricular expression of thrombin receptor also termed as protease-activated receptor-1 or PAR1, p-AKT (ser-473), p-50 NFκB and caspase-3 proteins. This study demonstrates beneficial effects of argatroban via improvement in cardiac functions and structural changes in STZ-induced DCM. These effects may be attributed through reduction in cardiac inflammation, fibrosis and apoptosis.
Collapse
Affiliation(s)
- Yogesh Bulani
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector-67, S.A.S. Nagar, Mohali, 160062, Punjab, India
| | - Krishnamoorthy Srinivasan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector-67, S.A.S. Nagar, Mohali, 160062, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector-67, S.A.S. Nagar, Mohali, 160062, Punjab, India.
| |
Collapse
|
22
|
Shan Z, Liu X, Chen Y, Wang M, Gao YR, Xu L, Dar WA, Lee CG, Elias JA, Castillo PD, Di Paola J, Ju C. Chitinase 3-like-1 promotes intrahepatic activation of coagulation through induction of tissue factor in mice. Hepatology 2018; 67:2384-2396. [PMID: 29251791 PMCID: PMC5992002 DOI: 10.1002/hep.29733] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 12/06/2017] [Accepted: 12/11/2017] [Indexed: 01/03/2023]
Abstract
Coagulation is a critical component in the progression of liver disease. Identification of key molecules involved in the intrahepatic activation of coagulation (IAOC) will be instrumental in the development of effective therapies against liver disease. Using a mouse model of concanavalin A (ConA)-induced hepatitis, in which IAOC plays an essential role in causing liver injury, we uncovered a procoagulant function of chitinase 3-like 1 (Chi3l1). Chi3l1 expression is dramatically elevated after ConA challenge, which is dependent on ConA-induced T cell activation and the resulting interferon γ and tumor necrosis factor α productions. Compared with wild-type mice, Chi3l1-/- mice show less IAOC, reduced tissue factor (TF) expression, and attenuated liver injury. Reconstituting Chi3l1-/- mice with recombinant TF triggers IAOC and augments liver injury. CONCLUSION Our data demonstrate that Chi3l1, through induction of TF via mitogen-activated protein kinase activation, promotes IAOC and tissue injury. (Hepatology 2018;67:2384-2396).
Collapse
Affiliation(s)
- Zhao Shan
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO, USA
| | - Xiaodong Liu
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO, USA
| | - Yuan Chen
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO, USA
| | - Meng Wang
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO, USA
| | - Yue Rachel Gao
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO, USA
| | - Liangguo Xu
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO, USA
| | - Wasim A. Dar
- Department of Surgery, UTHealth McGovern Medical School, Houston, TX, USA
| | - Chun Geun Lee
- Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, New Haven, CT, USA
| | - Jack Angel Elias
- Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, New Haven, CT, USA
- Division of Medicine and Biological Sciences, Warren Alpert School of Medicine, Brown University, Providence, Rhode Island, New Haven, CT, USA
| | - Pavel Davizon Castillo
- Department of Pediatric, School of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Jorge Di Paola
- Department of Pediatric, School of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Cynthia Ju
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO, USA
- Integrated Department of Immunology, University of Colorado Denver Aurora, CO, USA
| |
Collapse
|
23
|
Kopec AK, Abrahams SR, Thornton S, Palumbo JS, Mullins ES, Divanovic S, Weiler H, Owens AP, Mackman N, Goss A, van Ryn J, Luyendyk JP, Flick MJ. Thrombin promotes diet-induced obesity through fibrin-driven inflammation. J Clin Invest 2017; 127:3152-3166. [PMID: 28737512 DOI: 10.1172/jci92744] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 06/02/2017] [Indexed: 02/06/2023] Open
Abstract
Obesity promotes a chronic inflammatory and hypercoagulable state that drives cardiovascular disease, type 2 diabetes, fatty liver disease, and several cancers. Elevated thrombin activity underlies obesity-linked thromboembolic events, but the mechanistic links between the thrombin/fibrin(ogen) axis and obesity-associated pathologies are incompletely understood. In this work, immunohistochemical studies identified extravascular fibrin deposits within white adipose tissue and liver as distinct features of mice fed a high-fat diet (HFD) as well as obese patients. Fibγ390-396A mice carrying a mutant form of fibrinogen incapable of binding leukocyte αMβ2-integrin were protected from HFD-induced weight gain and elevated adiposity. Fibγ390-396A mice had markedly diminished systemic, adipose, and hepatic inflammation with reduced macrophage counts within white adipose tissue, as well as near-complete protection from development of fatty liver disease and glucose dysmetabolism. Homozygous thrombomodulin-mutant ThbdPro mice, which have elevated thrombin procoagulant function, gained more weight and developed exacerbated fatty liver disease when fed a HFD compared with WT mice. In contrast, treatment with dabigatran, a direct thrombin inhibitor, limited HFD-induced obesity development and suppressed progression of sequelae in mice with established obesity. Collectively, these data provide proof of concept that targeting thrombin or fibrin(ogen) may limit pathologies in obese patients.
Collapse
Affiliation(s)
- Anna K Kopec
- Department of Pathobiology and Diagnostic Investigation, Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | | | | | | | | | - Senad Divanovic
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, USA
| | - Hartmut Weiler
- Department of Physiology, Blood Center of Wisconsin, Milwaukee, Wisconsin, USA
| | - A Phillip Owens
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Nigel Mackman
- Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ashley Goss
- Department of Cardiometabolic Disease Research, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
| | - Joanne van Ryn
- Department of Cardiometabolic Disease Research, Boehringer Ingelheim Pharma GmbH, Biberach, Germany
| | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | | |
Collapse
|
24
|
Nault R, Fader KA, Kopec AK, Harkema JR, Zacharewski TR, Luyendyk JP. From the Cover: Coagulation-Driven Hepatic Fibrosis Requires Protease Activated Receptor-1 (PAR-1) in a Mouse Model of TCDD-Elicited Steatohepatitis. Toxicol Sci 2016; 154:381-391. [PMID: 27613713 DOI: 10.1093/toxsci/kfw175] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Emerging evidence supports a role for environmental chemical exposure in the pathology of non-alcoholic fatty liver disease (NAFLD), a disease process tightly linked to increased activity of the blood coagulation cascade. Exposure of C57BL/6 mice to the persistent environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) recapitulates features of the NAFLD spectrum, including steatosis, hepatic injury, inflammation, and fibrosis. We assessed coagulation cascade activation, and determined the role of the thrombin receptor protease activated receptor-1 (PAR-1) in experimental TCDD-elicited NAFLD. Chronic exposure to TCDD (30 µg/kg every 4 days for 28 days) was associated with intrahepatic coagulation, indicated by increased plasma thrombin-antithrombin levels and hepatic fibrin(ogen) deposition. PAR-1 deficiency diminished TCDD-elicited body weight loss and relative liver weight was reduced in TCDD-exposed PAR-1-/- mice compared with TCDD-exposed wild-type mice. PAR-1 deficiency did not affect TCDD-induced hepatic steatosis or hepatocellular injury, as indicated by serum alanine aminotransferase activity. Despite a lack of effect on these 2 features of NAFLD pathology, PAR-1 deficiency was associated with a reduction in hepatic inflammation evident in liver histopathology, and reflected by a reduction in serum levels of the proinflammatory cytokine interleukin-6. Moreover, TCDD-driven hepatic collagen deposition was markedly reduced in PAR-1-deficient mice. These results indicate that experimental TCDD-elicited steatohepatitis is associated with coagulation cascade activation and PAR-1-driven hepatic inflammation and fibrosis.
Collapse
Affiliation(s)
- Rance Nault
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan.,Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan
| | - Kelly A Fader
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan.,Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan
| | - Anna K Kopec
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan.,Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - Jack R Harkema
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - Timothy R Zacharewski
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan.,Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan
| | - James P Luyendyk
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan; .,Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| |
Collapse
|
25
|
Kopec AK, Joshi N, Luyendyk JP. Role of hemostatic factors in hepatic injury and disease: animal models de-liver. J Thromb Haemost 2016; 14:1337-49. [PMID: 27060337 PMCID: PMC5091081 DOI: 10.1111/jth.13327] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Indexed: 12/14/2022]
Abstract
Chronic liver damage is associated with unique changes in the hemostatic system. Patients with liver disease often show a precariously rebalanced hemostatic system, which is easily tipped towards bleeding or thrombotic complications by otherwise benign stimuli. In addition, some clinical studies have shown that hemostatic system components contribute to the progression of liver disease. There is a strong basic science foundation for clinical studies with this particular focus. Chronic and acute liver disease can be modeled in rodents and large animals with a variety of approaches, which span chronic exposure to toxic xenobiotics, diet-induced obesity, and surgical intervention. These experimental approaches have now provided strong evidence that, in addition to perturbations in hemostasis caused by liver disease, elements of the hemostatic system have powerful effects on the progression of experimental liver toxicity and disease. In this review, we cover the basis of the animal models that are most often utilized to assess the impact of the hemostatic system on liver disease, and highlight the role that coagulation proteases and their targets play in experimental liver toxicity and disease, emphasizing key similarities and differences between models. The need to characterize hemostatic changes in existing animal models and to develop novel animal models recapitulating the coagulopathy of chronic liver disease is highlighted. Finally, we emphasize the continued need to translate knowledge derived from highly applicable animal models to improve our understanding of the reciprocal interaction between liver disease and the hemostatic system in patients.
Collapse
Affiliation(s)
- Anna K. Kopec
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, Michigan 48824
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| | - Nikita Joshi
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan 48824
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| | - James P. Luyendyk
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, Michigan 48824
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan 48824
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| |
Collapse
|
26
|
Kopec AK, Luyendyk JP. Role of Fibrin(ogen) in Progression of Liver Disease: Guilt by Association? Semin Thromb Hemost 2016; 42:397-407. [PMID: 27144445 DOI: 10.1055/s-0036-1579655] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Strong experimental evidence indicates that components of the hemostatic system, including thrombin, exacerbate diverse features of experimental liver disease. Clinical studies have also begun to address this connection and some studies have suggested that anticoagulants can improve outcome in patients with liver disease. Among the evidence of coagulation cascade activation in models of liver injury and disease is the frequent observation of thrombin-driven hepatic fibrin(ogen) deposition. Indeed, hepatic fibrin(ogen) deposition has long been recognized as a consequence of hepatic injury. Although commonly inferred as pathologic due to protective effects of anticoagulants in mouse models, the role of fibrin(ogen) in acute liver injury and chronic liver disease may not be universally detrimental. The localization of hepatic fibrin(ogen) deposits within the liver is connected to the disease stimulus and in animal models of liver toxicity and chronic disease, fibrin(ogen) deposition may not always be synonymous with large vessel thrombosis. Here, we provide a balanced review of the experimental evidence supporting a direct connection between fibrin(ogen) and liver injury/disease pathogenesis, and suggest a path forward bridging experimental and clinical research to improve our knowledge on the nature and function of fibrin(ogen) in liver disease.
Collapse
Affiliation(s)
- Anna K Kopec
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| |
Collapse
|
27
|
Platelets and protease-activated receptor-4 contribute to acetaminophen-induced liver injury in mice. Blood 2015; 126:1835-43. [PMID: 26179083 DOI: 10.1182/blood-2014-09-598656] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 07/13/2015] [Indexed: 12/15/2022] Open
Abstract
Acetaminophen (APAP)-induced liver injury in humans is associated with robust coagulation cascade activation and thrombocytopenia. However, it is not known whether coagulation-driven platelet activation participates in APAP hepatotoxicity. Here, we found that APAP overdose in mice caused liver damage accompanied by significant thrombocytopenia and accumulation of platelets in the liver. These changes were attenuated by administration of the direct thrombin inhibitor lepirudin. Platelet depletion with an anti-CD41 antibody also significantly reduced APAP-mediated liver injury and thrombin generation, indicated by the concentration of thrombin-antithrombin (TAT) complexes in plasma. Compared with APAP-treated wild-type mice, biomarkers of hepatocellular and endothelial damage, plasma TAT concentration, and hepatic platelet accumulation were reduced in mice lacking protease-activated receptor (PAR)-4, which mediates thrombin signaling in mouse platelets. However, selective hematopoietic cell PAR-4 deficiency did not affect APAP-induced liver injury or plasma TAT levels. These results suggest that interconnections between coagulation and hepatic platelet accumulation promote APAP-induced liver injury, independent of platelet PAR-4 signaling. Moreover, the results highlight a potential contribution of nonhematopoietic cell PAR-4 signaling to APAP hepatotoxicity.
Collapse
|
28
|
Shih DM, Yu JM, Vergnes L, Dali-Youcef N, Champion MD, Devarajan A, Zhang P, Castellani LW, Brindley DN, Jamey C, Auwerx J, Reddy ST, Ford DA, Reue K, Lusis AJ. PON3 knockout mice are susceptible to obesity, gallstone formation, and atherosclerosis. FASEB J 2015; 29:1185-97. [PMID: 25477283 PMCID: PMC4396607 DOI: 10.1096/fj.14-260570] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 11/07/2014] [Indexed: 11/11/2022]
Abstract
We report the engineering and characterization of paraoxonase-3 knockout mice (Pon3KO). The mice were generally healthy but exhibited quantitative alterations in bile acid metabolism and a 37% increased body weight compared to the wild-type mice on a high fat diet. PON3 was enriched in the mitochondria-associated membrane fraction of hepatocytes. PON3 deficiency resulted in impaired mitochondrial respiration, increased mitochondrial superoxide levels, and increased hepatic expression of inflammatory genes. PON3 deficiency did not influence atherosclerosis development on an apolipoprotein E null hyperlipidemic background, but it did lead to a significant 60% increase in atherosclerotic lesion size in Pon3KO mice on the C57BL/6J background when fed a cholate-cholesterol diet. On the diet, the Pon3KO had significantly increased plasma intermediate-density lipoprotein/LDL cholesterol and bile acid levels. They also exhibited significantly elevated levels of hepatotoxicity markers in circulation, a 58% increase in gallstone weight, a 40% increase in hepatic cholesterol level, and increased mortality. Furthermore, Pon3KO mice exhibited decreased hepatic bile acid synthesis and decreased bile acid levels in the small intestine compared with wild-type mice. Our study suggests a role for PON3 in the metabolism of lipid and bile acid as well as protection against atherosclerosis, gallstone disease, and obesity.
Collapse
Affiliation(s)
- Diana M Shih
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Janet M Yu
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Laurent Vergnes
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Nassim Dali-Youcef
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Matthew D Champion
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Asokan Devarajan
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Peixiang Zhang
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Lawrence W Castellani
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - David N Brindley
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Carole Jamey
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Johan Auwerx
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Srinivasa T Reddy
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - David A Ford
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Karen Reue
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Aldons J Lusis
- *Division of Cardiology, Department of Medicine, Department of Microbiology, Immunology, and Molecular Genetics, Department of Human Genetics, Department of Molecular and Medical Pharmacology, and Department of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA; IGBMC, Illkirch and Hôpitaux Universitaires de Strasbourg, and **Laboratoire de Toxicologie, Universitaires de Strasbourg, Strasbourg, France; Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, St. Louis University School of Medicine, St. Louis, Missouri, USA; University of Alberta, Edmonton, Alberta, Canada; and Laboratory for Integrative and Systems Physiology, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
29
|
Ruf W, Samad F. Tissue factor pathways linking obesity and inflammation. Hamostaseologie 2015; 35:279-83. [PMID: 25623940 DOI: 10.5482/hamo-14-11-0068] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 01/13/2015] [Indexed: 01/26/2023] Open
Abstract
Obesity is a major cause for a spectrum of metabolic syndrome-related diseases that include insulin resistance, type 2 diabetes, and steatosis of the liver. Inflammation elicited by macrophages and other immune cells contributes to the metabolic abnormalities in obesity. In addition, coagulation activation following tissue factor (TF) upregulation in adipose tissue is frequently found in obese patients and particularly associated with diabetic complications. Genetic and pharmacological evidence indicates that TF makes significant contributions to the development of the metabolic syndrome by signaling through G protein-coupled protease activated receptors (PARs). Adipocyte TF-PAR2 signaling contributes to diet-induced obesity by decreasing metabolism and energy expenditure, whereas hematopoietic TF-PAR2 signaling is a major cause for adipose tissue inflammation, hepatic steatosis and inflammation, as well as insulin resistance. In the liver of mice on a high fat diet, PAR2 signaling increases transcripts of key regulators of gluconeogenesis, lipogenesis and inflammatory cytokines. Increased markers of hepatic gluconeogenesis correlate with decreased activation of AMP-activated protein kinase (AMPK), a known regulator of these pathways and a target for PAR2 signaling. Clinical markers of a TF-induced prothrombotic state may thus indicate a risk in obese patient for developing complications of the metabolic syndrome.
Collapse
Affiliation(s)
- W Ruf
- Wolfram Ruf, M.D., Professor, Department of Immunology and Microbial Science, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, Mail stop: SP258, Tel. 858/784-2748, Fax -8480, E-mail: ,
| | | |
Collapse
|
30
|
Hematopoietic tissue factor-protease-activated receptor 2 signaling promotes hepatic inflammation and contributes to pathways of gluconeogenesis and steatosis in obese mice. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:524-35. [PMID: 25476527 DOI: 10.1016/j.ajpath.2014.10.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 10/08/2014] [Accepted: 10/14/2014] [Indexed: 12/19/2022]
Abstract
Failure to inhibit hepatic gluconeogenesis is a major mechanism contributing to fasting hyperglycemia in type 2 diabetes and, along with steatosis, is the hallmark of hepatic insulin resistance. Obesity is associated with chronic inflammation in multiple tissues, and hepatic inflammation is mechanistically linked to both steatosis and hepatic insulin resistance. Here, we delineate a role for coagulation signaling via tissue factor (TF) and proteinase-activated receptor 2 (PAR2) in obesity-mediated hepatic inflammation, steatosis, and gluconeogenesis. In diet-induced obese mice, TF tail signaling independent of PAR2 drives CD11b(+)CD11c(+) hepatic macrophage recruitment, and TF-PAR2 signaling contributes to the accumulation of hepatic CD8(+) T cells. Transcripts of key pathways of gluconeogenesis, lipogenesis, and inflammatory cytokines were reduced in high-fat diet-fed mice that lack the cytoplasmic domain of TF (F3) (TF(ΔCT)) or that are deficient in PAR2 (F2rl1), as well as by pharmacological inhibition of TF-PAR2 signaling in diet-induced obese mice. These gluconeogenic, lipogenic, and inflammatory pathway transcripts were similarly reduced in response to genetic ablation or pharmacological inhibition of TF-PAR2 signaling in hematopoietic cells and were mechanistically associated with activation of AMP-activated protein kinase (AMPK). These findings indicate that hematopoietic TF-PAR2 signaling plays a pivotal role in the hepatic inflammatory responses, steatosis, and hepatic insulin resistance that lead to systemic insulin resistance and type 2 diabetes in obesity.
Collapse
|
31
|
Kopec AK, Luyendyk JP. Coagulation in liver toxicity and disease: role of hepatocyte tissue factor. Thromb Res 2014; 133 Suppl 1:S57-9. [PMID: 24759146 DOI: 10.1016/j.thromres.2014.03.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The liver is the primary source of a number of circulating coagulation factors, and acute liver injury and chronic liver disease are each associated with alterations in blood coagulation. Current views of the connection between liver injury and coagulation extend beyond the impact of liver disease on synthesis of coagulation factors to include a role for coagulation factor activity in the initiation and progression of liver disease. Mechanisms of coagulation initiation in liver disease are not completely understood. Compared to other tissues, liver expresses very low levels of tissue factor (TF). Recent studies indicate that expression of TF by hepatocytes comprises the majority of liver procoagulant activity, and that hepatocyte TF activates coagulation induced by liver injury. This review will briefly cover the expression and regulation of TF by hepatocytes, the role of TF in coagulation triggered by liver toxicity, and the contribution of coagulation activity to the progression of liver disease.
Collapse
Affiliation(s)
- Anna K Kopec
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI 48824
| | - James P Luyendyk
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI 48824.
| |
Collapse
|
32
|
Kopec AK, Joshi N, Towery KL, Kassel KM, Sullivan BP, Flick MJ, Luyendyk JP. Thrombin inhibition with dabigatran protects against high-fat diet-induced fatty liver disease in mice. J Pharmacol Exp Ther 2014; 351:288-97. [PMID: 25138021 PMCID: PMC4201275 DOI: 10.1124/jpet.114.218545] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 08/13/2014] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of obesity and metabolic syndrome. Robust coagulation cascade activation is common in obese patients with NAFLD. We identified a critical temporal relationship between thrombin generation and the manifestation of hepatic steatosis, inflammation, and injury in C57BL/6J mice fed a high-fat diet (HFD) for 1, 2, and 3 months. Mice fed a HFD exhibited dramatic increases in hepatocellular injury and inflammation over time. Hepatic fibrin deposition preceded an increase in serum alanine aminotransferase, and the most dramatic changes in liver histopathology occurred in conjunction with a detectable increase in plasma thrombin-antithrombin levels at 3 months. To directly determine whether thrombin activity promotes NAFLD pathogenesis, mice were fed a HFD and simultaneously treated with the direct thrombin inhibitor dabigatran etexilate for 3 months. Notably, dabigatran treatment significantly reduced hepatic fibrin deposition, hepatic inflammation, hepatocellular injury, and steatosis in mice fed a HFD. Of interest, dabigatran treatment also significantly attenuated HFD-induced body weight gain. Gene expression analysis suggested that thrombin potentially drives NAFLD pathogenesis by altering the expression of genes associated with lipid metabolism and bile acid synthesis. Collectively, the results suggest that thrombin activity is central to HFD-induced body weight gain, liver injury, and inflammation and provide the proof-of-principle evidence that pharmacological thrombin inhibition could be effective in limiting NAFLD and associated pathologies.
Collapse
Affiliation(s)
- Anna K Kopec
- Department of Pathobiology & Diagnostic Investigation (A.K.K., K.L.T., J.P.L.), Department of Pharmacology & Toxicology (N.J.), Michigan State University, East Lansing, Michigan; Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (K.M.K., B.P.S.); and Cancer and Blood Diseases Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, Ohio (M.J.F.)
| | - Nikita Joshi
- Department of Pathobiology & Diagnostic Investigation (A.K.K., K.L.T., J.P.L.), Department of Pharmacology & Toxicology (N.J.), Michigan State University, East Lansing, Michigan; Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (K.M.K., B.P.S.); and Cancer and Blood Diseases Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, Ohio (M.J.F.)
| | - Keara L Towery
- Department of Pathobiology & Diagnostic Investigation (A.K.K., K.L.T., J.P.L.), Department of Pharmacology & Toxicology (N.J.), Michigan State University, East Lansing, Michigan; Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (K.M.K., B.P.S.); and Cancer and Blood Diseases Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, Ohio (M.J.F.)
| | - Karen M Kassel
- Department of Pathobiology & Diagnostic Investigation (A.K.K., K.L.T., J.P.L.), Department of Pharmacology & Toxicology (N.J.), Michigan State University, East Lansing, Michigan; Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (K.M.K., B.P.S.); and Cancer and Blood Diseases Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, Ohio (M.J.F.)
| | - Bradley P Sullivan
- Department of Pathobiology & Diagnostic Investigation (A.K.K., K.L.T., J.P.L.), Department of Pharmacology & Toxicology (N.J.), Michigan State University, East Lansing, Michigan; Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (K.M.K., B.P.S.); and Cancer and Blood Diseases Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, Ohio (M.J.F.)
| | - Matthew J Flick
- Department of Pathobiology & Diagnostic Investigation (A.K.K., K.L.T., J.P.L.), Department of Pharmacology & Toxicology (N.J.), Michigan State University, East Lansing, Michigan; Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (K.M.K., B.P.S.); and Cancer and Blood Diseases Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, Ohio (M.J.F.)
| | - James P Luyendyk
- Department of Pathobiology & Diagnostic Investigation (A.K.K., K.L.T., J.P.L.), Department of Pharmacology & Toxicology (N.J.), Michigan State University, East Lansing, Michigan; Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (K.M.K., B.P.S.); and Cancer and Blood Diseases Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, Ohio (M.J.F.)
| |
Collapse
|
33
|
Lopez M, Kopec AK, Joshi N, Geddings JE, Cline H, Towery KL, Rockwell CE, Mackman N, Luyendyk JP. Fas-induced apoptosis increases hepatocyte tissue factor procoagulant activity in vitro and in vivo. Toxicol Sci 2014; 141:453-64. [PMID: 25015658 DOI: 10.1093/toxsci/kfu139] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Hepatocyte (HPC) apoptosis occurs in association with hepatotoxic responses and chronic liver disease, and is coupled to activation of the blood coagulation cascade. HPCs have been shown to express tissue factor (TF), the primary activator of blood coagulation, in a form that lacks procoagulant activity. In this study, we determined the effect of inducing HPC apoptosis on the procoagulant activity of TF. Treatment of primary mouse HPCs with the Fas death receptor agonist (anti-CD95 antibody, Jo2) triggered apoptosis as shown by cleavage of caspase-3, increased caspase-3 proteolytic activity, and cell surface exposure of phosphatidylserine (PS). Jo2-induced apoptosis significantly increased TF-dependent factor Xa generation by HPCs. Moreover, Jo2 treatment was associated with increased levels of microparticle-associated TF procoagulant activity in the culture medium. Pretreatment with a caspase-3 inhibitor significantly reduced Jo2-induced HPC TF activity and prevented the increase in microparticle-associated TF procoagulant activity. Application of the high-affinity PS-binding protein lactadherin inhibited TF-dependent factor Xa generation by Jo2-treated HPCs and dramatically reduced microparticle-associated TF procoagulant activity. Treatment of wild-type mice with a sublethal dose of Jo2 was associated with a robust increase in the activation of coagulation as measured by plasma thrombin-antithrombin (TAT) levels; whereas mice with liver-specific TF deficiency had significantly lower TAT levels. Overall, the results indicate that Fas-initiated, caspase-3-dependent HPC apoptosis increases TF procoagulant activity through a mechanism involving PS externalization. This suggests that activation of liver TF likely contributes to the procoagulant state associated with HPC apoptosis in liver toxicity and disease.
Collapse
Affiliation(s)
- Michelle Lopez
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, 253 Food Safety and Toxicology Bldg, East Lansing, Michigan 48824
| | - Anna K Kopec
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, 253 Food Safety and Toxicology Bldg, East Lansing, Michigan 48824
| | - Nikita Joshi
- Department of Pharmacology and Toxicology, Michigan State University, B346 Life Sciences, Michigan State University, East Lansing, Michigan 48824
| | - Julia E Geddings
- Department of Medicine, University of North Carolina at Chapel Hill, 98 Manning Dr. Chapel Hill, North Carolina 27599
| | - Holly Cline
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, 253 Food Safety and Toxicology Bldg, East Lansing, Michigan 48824
| | - Keara L Towery
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, 253 Food Safety and Toxicology Bldg, East Lansing, Michigan 48824
| | - Cheryl E Rockwell
- Department of Pharmacology and Toxicology, Michigan State University, B346 Life Sciences, Michigan State University, East Lansing, Michigan 48824
| | - Nigel Mackman
- Department of Medicine, University of North Carolina at Chapel Hill, 98 Manning Dr. Chapel Hill, North Carolina 27599
| | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, 253 Food Safety and Toxicology Bldg, East Lansing, Michigan 48824
| |
Collapse
|
34
|
Abstract
Clinical and epidemiological studies support a connection between obesity and thrombosis, involving elevated expression of the prothrombotic molecules plasminogen activator inhibitor-1 and tissue factor (TF) and increased platelet activation. Cardiovascular diseases and metabolic syndrome-associated disorders, including obesity, insulin resistance, type 2 diabetes, and hepatic steatosis, involve inflammation elicited by infiltration and activation of immune cells, particularly macrophages, into adipose tissue. Although TF has been clearly linked to a procoagulant state in obesity, emerging genetic and pharmacologic evidence indicate that TF signaling via G protein-coupled protease-activated receptors (PAR2, PAR1) additionally drives multiple aspects of the metabolic syndrome. TF-PAR2 signaling in adipocytes contributes to diet-induced obesity by decreasing metabolism and energy expenditure, whereas TF-PAR2 signaling in hematopoietic and myeloid cells drives adipose tissue inflammation, hepatic steatosis, and insulin resistance. TF-initiated coagulation leading to thrombin-PAR1 signaling also contributes to diet-induced hepatic steatosis and inflammation in certain models. Thus, in obese patients, clinical markers of a prothrombotic state may indicate a risk for the development of complications of the metabolic syndrome. Furthermore, TF-induced signaling could provide new therapeutic targets for drug development at the intersection between obesity, inflammation, and thrombosis.
Collapse
|
35
|
Coagulation and coagulation signalling in fibrosis. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1018-27. [PMID: 23298546 DOI: 10.1016/j.bbadis.2012.12.013] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Revised: 12/20/2012] [Accepted: 12/22/2012] [Indexed: 12/29/2022]
Abstract
Following tissue injury, a complex and coordinated wound healing response comprising coagulation, inflammation, fibroproliferation and tissue remodelling has evolved to nullify the impact of the original insult and reinstate the normal physiological function of the affected organ. Tissue fibrosis is thought to result from a dysregulated wound healing response as a result of continual local injury or impaired control mechanisms. Although the initial insult is highly variable for different organs, in most cases, uncontrolled or sustained activation of mesenchymal cells into highly synthetic myofibroblasts leads to the excessive deposition of extracellular matrix proteins and eventually loss of tissue function. Coagulation was originally thought to be an acute and transient response to tissue injury, responsible primarily for promoting haemostasis by initiating the formation of fibrin plugs to enmesh activated platelets within the walls of damaged blood vessels. However, the last 20years has seen a major re-evaluation of the role of the coagulation cascade following tissue injury and there is now mounting evidence that coagulation plays a critical role in orchestrating subsequent inflammatory and fibroproliferative responses during normal wound healing, as well as in a range of pathological contexts across all major organ systems. This review summarises our current understanding of the role of coagulation and coagulation initiated signalling in the response to tissue injury, as well as the contribution of uncontrolled coagulation to fibrosis of the lung, liver, kidney and heart. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
|
36
|
Therapeutic administration of the direct thrombin inhibitor argatroban reduces hepatic inflammation in mice with established fatty liver disease. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:1287-95. [PMID: 22841818 DOI: 10.1016/j.ajpath.2012.06.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 05/14/2012] [Accepted: 06/18/2012] [Indexed: 02/08/2023]
Abstract
Thrombin generation is increased in patients with nonalcoholic fatty liver disease (NAFLD) and in mouse models of diet-induced obesity. Deficiency in the thrombin receptor protease activated receptor-1 reduces hepatic inflammation and steatosis in mice fed a Western diet. However, it is currently unclear whether thrombin inhibitors can modify the pathogenesis of established NAFLD. We tested the hypothesis that thrombin inhibition could reverse hepatic steatosis and inflammation in mice with established diet-induced NAFLD. Low-density lipoprotein receptor-deficient LDLr(-/-) mice were fed a control diet or a Western diet for 19 weeks. Mice were given the direct thrombin inhibitor argatroban ∼15 mg/kg/day or its vehicle via a miniosmotic pump for the final 4 weeks of the study. Argatroban administration significantly reduced hepatic proinflammatory cytokine expression and reduced macrophage and neutrophil accumulation in livers of mice fed a Western diet. Argatroban did not significantly impact hepatic steatosis, as indicated by histopathology, Oil Red O staining, and hepatic triglyceride levels. Argatroban reduced serum triglyceride and cholesterol levels in mice fed a Western diet. Argatroban reduced both α-smooth muscle actin expression and Type 1 collagen mRNA levels in livers of mice fed a Western diet, indicating reduced activation of hepatic stellate cells. This study indicates that therapeutic intervention with a thrombin inhibitor attenuates hepatic inflammation and several profibrogenic changes in mice fed a Western diet.
Collapse
|
37
|
Luo W, Öhman M, Wright A, Kamrudin S, Wang H, Guo C, Eitzman D. Steatohepatitis and vascular thrombosis in apolipoprotein e deficient mice. Thromb Res 2012; 129:e166-7. [PMID: 22326191 DOI: 10.1016/j.thromres.2012.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 01/04/2012] [Accepted: 01/16/2012] [Indexed: 11/16/2022]
|
38
|
|