1
|
Dai Q, Ain Q, Seth N, Rooney M, Zipprich A. Liver sinusoidal endothelial cells: Friend or foe in metabolic dysfunction- associated steatotic liver disease/metabolic dysfunction-associated steatohepatitis. Dig Liver Dis 2025; 57:493-503. [PMID: 39904692 DOI: 10.1016/j.dld.2025.01.189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/27/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the predominant liver disease and is becoming the paramount contributor to end-stage liver disease and liver-related deaths. Liver sinusoidal endothelial cells (LSECs) located between the hepatic parenchyma and blood from viscera and gastrointestinal tract are the gatekeepers for the hepatic microenvironment and normal function. In normal physiological conditions, LSECs govern the substance exchange between hepatic parenchyma and blood through dynamic regulation of fenestration and maintain the quiescent state of Kupffer cells (KCs) and hepatic stellate cells. In MASLD, lipotoxicity, insulin resistance, gastrointestinal microbiota dysbiosis, and mechanical compression caused by fat-laden hepatocytes result in LSECs capillarization and dysfunction. The altered LSECs progressively shift from healer to injurer, exacerbating liver inflammation and advancing liver fibrosis. This review focuses on the deteriorative roles of LSECs and related molecular mechanisms involved in MASLD and their contribution to metabolic dysfunction-associated steatohepatitis (MASH) and liver fibrosis development and progression. Furthermore, in this review, we propose that targeting LSECs dysfunction is a prospective therapeutic strategy to restore the physiological function of LSECs and mitigate MASLD progression.
Collapse
Affiliation(s)
- Qingqing Dai
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, 07747, Jena, Thuringia, Germany
| | - Quratul Ain
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, 07747, Jena, Thuringia, Germany
| | - Navodita Seth
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, 07747, Jena, Thuringia, Germany
| | - Michael Rooney
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, 07747, Jena, Thuringia, Germany
| | - Alexander Zipprich
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, 07747, Jena, Thuringia, Germany.
| |
Collapse
|
2
|
Ning M, Lu D, Liang D, Ren PG. Single-cell RNA sequencing advances in revealing the development and progression of MASH: the identifications and interactions of non-parenchymal cells. Front Mol Biosci 2025; 12:1513993. [PMID: 40201243 PMCID: PMC11976672 DOI: 10.3389/fmolb.2025.1513993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 03/05/2025] [Indexed: 04/10/2025] Open
Abstract
Developing drugs for the treatment of Metabolic Associated Steatohepatitis (MASH) has always been a significant challenge. Researchers have been dedicated to exploring drugs and therapeutic strategies to alleviate disease progression, but treatments remain limited. This is partly due to the complexity of the pathophysiological processes, and inadequate knowledge of the cellular and molecular mechanisms in MASH. Especially, the liver non-parenchymal cells (NPCs) like Kupffer cells, hepatic stellate cells and sinusoidal endothelial cells which play critical roles in live function, immune responses, fibrosis and disease progression. Deciphering how these cells function in MASH, would help understand the pathophysiological processes and find potential drug targets. In recent years, new technologies have been developed for single-cell transcriptomic sequencing, making cell-specific transcriptome profiling a reality in healthy and diseased livers. In this review, we discussed how the use of single-cell transcriptomic sequencing provided us with an in-depth understanding of the heterogeneous, cellular interactions among non-parenchymal cells and tried to highlight recent discoveries in MASH by this technology. It is hoped that the summarized features and markers of various subclusters in this review could provide a technical reference for further experiments and a theoretical basis for clinical applications.
Collapse
Affiliation(s)
- Meng Ning
- Department of Endocrinology, Peking University Shenzhen Hospital, Shenzhen, China
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Donghui Lu
- Department of Endocrinology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Dong Liang
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Pei-Gen Ren
- Center for Cancer Immunology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
3
|
Roser LA, Sakellariou C, Lindstedt M, Neuhaus V, Dehmel S, Sommer C, Raasch M, Flandre T, Roesener S, Hewitt P, Parnham MJ, Sewald K, Schiffmann S. IL-2-mediated hepatotoxicity: knowledge gap identification based on the irAOP concept. J Immunotoxicol 2024; 21:2332177. [PMID: 38578203 DOI: 10.1080/1547691x.2024.2332177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 03/13/2024] [Indexed: 04/06/2024] Open
Abstract
Drug-induced hepatotoxicity constitutes a major reason for non-approval and post-marketing withdrawal of pharmaceuticals. In many cases, preclinical models lack predictive capacity for hepatic damage in humans. A vital concern is the integration of immune system effects in preclinical safety assessment. The immune-related Adverse Outcome Pathway (irAOP) approach, which is applied within the Immune Safety Avatar (imSAVAR) consortium, presents a novel method to understand and predict immune-mediated adverse events elicited by pharmaceuticals and thus targets this issue. It aims to dissect the molecular mechanisms involved and identify key players in drug-induced side effects. As irAOPs are still in their infancy, there is a need for a model irAOP to validate the suitability of this tool. For this purpose, we developed a hepatotoxicity-based model irAOP for recombinant human IL-2 (aldesleukin). Besides producing durable therapeutic responses against renal cell carcinoma and metastatic melanoma, the boosted immune activation upon IL-2 treatment elicits liver damage. The availability of extensive data regarding IL-2 allows both the generation of a comprehensive putative irAOP and to validate the predictability of the irAOP with clinical data. Moreover, IL-2, as one of the first cancer immunotherapeutics on the market, is a blueprint for various biological and novel treatment regimens that are under investigation today. This review provides a guideline for further irAOP-directed research in immune-mediated hepatotoxicity.
Collapse
Affiliation(s)
- Luise A Roser
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
| | | | - Malin Lindstedt
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Vanessa Neuhaus
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Preclinical Pharmacology and In-Vitro Toxicology, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Member of the Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Hannover, Germany
| | - Susann Dehmel
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Preclinical Pharmacology and In-Vitro Toxicology, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Member of the Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Hannover, Germany
| | - Charline Sommer
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Preclinical Pharmacology and In-Vitro Toxicology, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Member of the Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Hannover, Germany
| | | | - Thierry Flandre
- Translational Medicine, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Sigrid Roesener
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - Michael J Parnham
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
- EpiEndo Pharmaceuticals ehf, Reykjavík, Iceland
| | - Katherina Sewald
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Preclinical Pharmacology and In-Vitro Toxicology, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Member of the Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Hannover, Germany
| | | |
Collapse
|
4
|
Gao J, Lan T, Kostallari E, Guo Y, Lai E, Guillot A, Ding B, Tacke F, Tang C, Shah VH. Angiocrine signaling in sinusoidal homeostasis and liver diseases. J Hepatol 2024; 81:543-561. [PMID: 38763358 PMCID: PMC11906189 DOI: 10.1016/j.jhep.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 05/21/2024]
Abstract
The hepatic sinusoids are composed of liver sinusoidal endothelial cells (LSECs), which are surrounded by hepatic stellate cells (HSCs) and contain liver-resident macrophages called Kupffer cells, and other patrolling immune cells. All these cells communicate with each other and with hepatocytes to maintain sinusoidal homeostasis and a spectrum of hepatic functions under healthy conditions. Sinusoidal homeostasis is disrupted by metabolites, toxins, viruses, and other pathological factors, leading to liver injury, chronic liver diseases, and cirrhosis. Alterations in hepatic sinusoids are linked to fibrosis progression and portal hypertension. LSECs are crucial regulators of cellular crosstalk within their microenvironment via angiocrine signaling. This review discusses the mechanisms by which angiocrine signaling orchestrates sinusoidal homeostasis, as well as the development of liver diseases. Here, we summarise the crosstalk between LSECs, HSCs, hepatocytes, cholangiocytes, and immune cells in health and disease and comment on potential novel therapeutic methods for treating liver diseases.
Collapse
Affiliation(s)
- Jinhang Gao
- Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Lan
- Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China; Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Enis Kostallari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Yangkun Guo
- Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Enjiang Lai
- Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Adrien Guillot
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Bisen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany.
| | - Chengwei Tang
- Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China.
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
5
|
Yang Y, Yu L, Sheng Z, Lin H, Weng Z, Song W, Cao B, Zhao Y, Gao Y, Ni S, Wang H, Ma T, Huang L, Sun C, Li J. The first selective VAP-1 inhibitor in China, TT-01025-CL: safety, tolerability, pharmacokinetics, and pharmacodynamics of single- and multiple-ascending doses. Front Pharmacol 2024; 15:1327008. [PMID: 38741586 PMCID: PMC11089243 DOI: 10.3389/fphar.2024.1327008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 04/09/2024] [Indexed: 05/16/2024] Open
Abstract
Introduction: TT-01025-CL is an oral, irreversible small molecule that potently inhibits vascular adhesion protein-1 (VAP-1) for the treatment of inflammation associated with non-alcoholic steatohepatitis (NASH). The objectives of this study were to evaluate the safety/tolerability, pharmacokinetics, and pharmacodynamics of TT-01025-CL, a VAP-1 inhibitor, in healthy Chinese volunteers. Methods: Double-blind, placebo-controlled, dose-escalation studies were conducted in subjects randomized to receive oral once-daily TT-01025-CL (ranges: 10-300 mg [single dose]; 20-100 mg for 7 days [multiple doses]) or placebo under fasting conditions. Safety and tolerability were monitored throughout the study. Pharmacokinetic (PK) parameters were determined using non-compartment analysis. The activity of semicarbazide-sensitive amine oxidase (SSAO)-specific amine oxidase and the accumulation of methylamine in plasma were evaluated as pharmacodynamic (PD) biomarkers. Results: A total of 36 (single-dose group) and 24 (multiple-dose group) subjects were enrolled in the study. No serious adverse events (AEs) were reported, and no subject discontinued due to an AE. All treatment-emergent adverse events (TEAEs) were mild and moderate in intensity. No dose-dependent increase in the intensity or frequency of events was observed. TT-01025-CL was rapidly absorbed after administration. In the single-ascending dose (SAD) study, median Tmax ranged from 0.5 to 2 h and mean t1/2z ranged from 2.09 to 4.39 h. PK was linear in the range of 100-300 mg. The mean Emax of methylamine ranged from 19.167 to 124.970 ng/mL, with mean TEmax ranging from 13.5 to 28.0 h. The complete inhibition (>90%) of SSAO activity was observed at 0.25-0.5 h post-dose and was maintained 48-168 h post-dose. In the multiple-ascending dose (MAD) study, a steady state was reached by day 5 in the 40 mg and 100 mg dose groups. Negligible accumulation was observed after repeated dosing. PK was linear in the range of 20-100 mg. Plasma methylamine appeared to plateau at doses of 20 mg and above, with mean Emax ranging from 124.142 to 156.070 ng/mL and mean TEmax ranging from 14.2 to 22.0 h on day 7. SSAO activity in plasma was persistently inhibited throughout the treatment period. No evident change in methylamine and SSAO activity was observed in the placebo groups. Conclusion: TT-01025-CL was safe and well-tolerated at a single dose of up to 300 mg and multiple doses of up to 100 mg once daily for 7 consecutive days. Absorption and elimination occurred rapidly in healthy volunteers. Linearity in plasma exposure was observed. TT-01025-CL inhibited SSAO activity rapidly and persistently in humans. The profile of TT-01025-CL demonstrates its suitability for further clinical development.
Collapse
Affiliation(s)
- Yuanxun Yang
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Lei Yu
- TransThera Sciences (Nanjing), Inc., Nanjing, China
| | - Zejuan Sheng
- TransThera Sciences (Nanjing), Inc., Nanjing, China
| | - Hui Lin
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zuyi Weng
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wei Song
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Bei Cao
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yu Zhao
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | | | - Shumao Ni
- TransThera Sciences (Nanjing), Inc., Nanjing, China
| | - Huimin Wang
- TransThera Sciences (Nanjing), Inc., Nanjing, China
| | - Tingting Ma
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Lei Huang
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Caixia Sun
- TransThera Sciences (Nanjing), Inc., Nanjing, China
| | - Juan Li
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
6
|
Lasagna A, Sacchi P. The ABC of Immune-Mediated Hepatitis during Immunotherapy in Patients with Cancer: From Pathogenesis to Multidisciplinary Management. Cancers (Basel) 2024; 16:795. [PMID: 38398187 PMCID: PMC10886483 DOI: 10.3390/cancers16040795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/29/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Immune-mediated hepatotoxicity (IMH) is not-so-rare complication during treatment with immune checkpoint inhibitors (ICIs). This narrative review aims to report the current knowledge on hepatic immune-related adverse events (irAEs) during immunotherapy from pathogenesis to multidisciplinary management. The majority of cases of IMH are asymptomatic and only a few patients may have clinical conditions. The severity of IMH is usually stratified according to Common Terminology for Clinical Adverse Events (CTCAE) criteria, but these scores may overestimate the clinical severity of IMH compared to the Drug-Induced Liver Injury Network (DILIN) scale. The differential diagnosis of IMH is challenging because the elevated liver enzymes can be due to a number of etiologies such as viral infection, autoimmune and metabolic diseases, liver metastases, biliary diseases, and other drugs. The cornerstones of IMH management are represented by withholding or delaying ICI administration and starting immunosuppressive therapy. A multidisciplinary team, including oncologists, hepatologists, internists, and emergency medicine physicians, is essential for the management of IMH.
Collapse
Affiliation(s)
- Angioletta Lasagna
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Paolo Sacchi
- Division of Infectious Diseases I, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
7
|
Santosh Nirmala S, Kayani K, Gliwiński M, Hu Y, Iwaszkiewicz-Grześ D, Piotrowska-Mieczkowska M, Sakowska J, Tomaszewicz M, Marín Morales JM, Lakshmi K, Marek-Trzonkowska NM, Trzonkowski P, Oo YH, Fuchs A. Beyond FOXP3: a 20-year journey unravelling human regulatory T-cell heterogeneity. Front Immunol 2024; 14:1321228. [PMID: 38283365 PMCID: PMC10811018 DOI: 10.3389/fimmu.2023.1321228] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/19/2023] [Indexed: 01/30/2024] Open
Abstract
The initial idea of a distinct group of T-cells responsible for suppressing immune responses was first postulated half a century ago. However, it is only in the last three decades that we have identified what we now term regulatory T-cells (Tregs), and subsequently elucidated and crystallized our understanding of them. Human Tregs have emerged as essential to immune tolerance and the prevention of autoimmune diseases and are typically contemporaneously characterized by their CD3+CD4+CD25high CD127lowFOXP3+ phenotype. It is important to note that FOXP3+ Tregs exhibit substantial diversity in their origin, phenotypic characteristics, and function. Identifying reliable markers is crucial to the accurate identification, quantification, and assessment of Tregs in health and disease, as well as the enrichment and expansion of viable cells for adoptive cell therapy. In our comprehensive review, we address the contributions of various markers identified in the last two decades since the master transcriptional factor FOXP3 was identified in establishing and enriching purity, lineage stability, tissue homing and suppressive proficiency in CD4+ Tregs. Additionally, our review delves into recent breakthroughs in innovative Treg-based therapies, underscoring the significance of distinct markers in their therapeutic utilization. Understanding Treg subsets holds the key to effectively harnessing human Tregs for immunotherapeutic approaches.
Collapse
Affiliation(s)
| | - Kayani Kayani
- Centre for Liver and Gastrointestinal Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Department of Academic Surgery, Queen Elizabeth Hospital, University of Birmingham, Birmingham, United Kingdom
- Department of Renal Surgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Mateusz Gliwiński
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Yueyuan Hu
- Center for Regenerative Therapies Dresden, Technical University Dresden, Dresden, Germany
| | | | | | - Justyna Sakowska
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Martyna Tomaszewicz
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Kavitha Lakshmi
- Center for Regenerative Therapies Dresden, Technical University Dresden, Dresden, Germany
| | | | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Ye Htun Oo
- Centre for Liver and Gastrointestinal Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Liver Transplant and Hepatobiliary Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Birmingham Advanced Cellular Therapy Facility, University of Birmingham, Birmingham, United Kingdom
- Centre for Rare Diseases, European Reference Network - Rare Liver Centre, Birmingham, United Kingdom
| | - Anke Fuchs
- Center for Regenerative Therapies Dresden, Technical University Dresden, Dresden, Germany
| |
Collapse
|
8
|
Lu Y, Guo X, Xu F, Wang F, Wu H, Bai Y, Li W, Zhang G, Yuan J, Pang Q. Protective effects of puerarin on liver tissue in Salmonella-infected chicks: a proteomic analysis. Poult Sci 2024; 103:103281. [PMID: 37992616 PMCID: PMC10700392 DOI: 10.1016/j.psj.2023.103281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/07/2023] [Accepted: 11/12/2023] [Indexed: 11/24/2023] Open
Abstract
Salmonella enterica is a zoonotic bacterium that not only causes serious economic losses to the livestock and poultry industries but also seriously endangers human health. Long-term indiscriminate use of antibiotics has led to drug resistance in Salmonella, and thus the identification of alternatives to antibiotics is crucial. In this study, the effects of puerarin on the S. enterica-infected chickens were investigated. A total of 360 chicks were randomly assigned as the control group (CON), the S. enterica group (S), and puerarin-treatment group (P). Chicks in the P group were fed the basal diet supplemented with 50 (P50), 100 (P100), 200 (P200), and 400 (P400) mg/kg puerarin, respectively. It was found that puerarin treatment markedly altered the serum activities of aspartate aminotransferase (AST), alanine transaminase (ALT), and superoxide dismutase (SOD), together with the malondialdehyde (MDA) and total antioxidant capacity (T-AOC) contents in the serum. The mRNA expression of IL-6, IL-1β, TNF-α, Bcl-2, and caspase-8 in the livers of S. enterica-infected chicks was increased after infection but significantly reduced after treatment with puerarin. Histologic analysis showed that puerarin effectively mitigated morphological damage in the liver caused by S. enterica. Proteomic analysis revealed that S. enterica infection led to metabolic disorders in the liver, resulting in oxidative stress, increased inflammation, and significantly elevated levels of hepatocellular carcinoma biomarkers. The findings of the filtered sequencing were verified by using quantitative PCR (qPCR). Treatment with 100 mg/mL puerarin thus effectively alleviated disordered liver metabolism, reduced inflammation and oxidative damage and significantly reduced the levels of hepatocellular carcinoma biomarkers in the liver. The results suggest that puerarin has the potential to replace antibiotics to control Salmonella infection in poultry and thus improve food safety.
Collapse
Affiliation(s)
- Yu Lu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China; College of Agriculture and Bioengineering, Heze University, Heze 274000, China
| | - Xiaotong Guo
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Fengge Xu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Fengyi Wang
- College of Agriculture and Bioengineering, Heze University, Heze 274000, China
| | - Hongsong Wu
- College of Agriculture and Bioengineering, Heze University, Heze 274000, China
| | - Yongjiang Bai
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Wenhua Li
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Guisheng Zhang
- College of Agriculture and Bioengineering, Heze University, Heze 274000, China
| | - Jinbao Yuan
- College of Agriculture and Bioengineering, Heze University, Heze 274000, China
| | - Quanhai Pang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China.
| |
Collapse
|
9
|
Alshehade S, Alshawsh MA, Murugaiyah V, Asif M, Alshehade O, Almoustafa H, Al Zarzour RH. The role of protein kinases as key drivers of metabolic dysfunction-associated fatty liver disease progression: New insights and future directions. Life Sci 2022; 305:120732. [PMID: 35760093 DOI: 10.1016/j.lfs.2022.120732] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 02/07/2023]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD), proposed in 2020 is a novel term for non-alcoholic fatty liver disease (NAFLD) which was coined for the first time in 1980. It is a leading cause of the most chronic liver disease and hepatic failure all over the world, and unfortunately, with no licensed drugs for treatment yet. The progress of the disease is driven by the triggered inflammatory process, oxidative stress, and insulin resistance in many pathways, starting with simple hepatic steatosis to non-alcoholic steatohepatitis, fibrosis, cirrhosis, and liver cancer. Protein kinases (PKs), such as MAPK, ErbB, PKC, PI3K/Akt, and mTOR, govern most of the pathological pathways by acting on various downstream key points in MAFLD and regulating both hepatic gluco- lipo-neogenesis and inflammation. Therefore, modulating the function of those potential protein kinases that are effectively involved in MAFLD might be a promising therapeutic approach for tackling this disease. In the current review, we have discussed the key role of protein kinases in the pathogenesis of MAFLD and performed a protein-protein interaction (PPI) network among the main proteins of each kinase pathway with MAFLD-related proteins to predict the most likely targets of the PKs in MAFLD. Moreover, we have reported the experimental, pre-clinical, and clinical data for the most recent investigated molecules that are activating p38-MAPK and AMPK proteins and inhibiting the other PKs to improve MAFLD condition by regulating oxidation and inflammation signalling.
Collapse
Affiliation(s)
- Salah Alshehade
- Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia; Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | | | - Vikneswaran Murugaiyah
- Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Muhammad Asif
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Punjab, Pakistan
| | - Omayma Alshehade
- Department of Paediatrics, Faculty of Medicine, Damascus University, Damascus, Syria
| | - Hassan Almoustafa
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Raghdaa Hamdan Al Zarzour
- Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia; Department of Pharmacology, Faculty of Pharmacy, Arab International University, Damascus, Syria.
| |
Collapse
|
10
|
Challenges and opportunities in achieving effective regulatory T cell therapy in autoimmune liver disease. Semin Immunopathol 2022; 44:461-474. [PMID: 35641679 PMCID: PMC9256571 DOI: 10.1007/s00281-022-00940-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/15/2022] [Indexed: 12/29/2022]
Abstract
Autoimmune liver diseases (AILD) include autoimmune hepatitis (AIH), primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC). These immune-mediated liver diseases involve a break down in peripheral self-tolerance with largely unknown aetiology. Regulatory T cells (Treg) are crucial in maintaining immunological tolerance. Hence, Treg immunotherapy is an attractive therapeutic option in AILD. Currently, AILD do not have a curative treatment option and patients take life-long immunosuppression or bile acids to control hepatic or biliary inflammation. Clinical investigations using good manufacturing practice (GMP) Treg in autoimmune liver disease have thus far demonstrated that Treg therapy is safe and that Treg migrate to inflamed liver tissue. For Treg immunotherapy to achieve efficacy in AILD, Treg must be retained within the liver and maintain their suppressive phenotype to dampen ongoing immune responses to hepatocytes and biliary epithelium. Therefore, therapeutic Treg subsets should be selected for tissue residency markers and maximal functionality. Optimisation of dosing regime and understanding longevity of Treg in vivo are critical to successful Treg therapy. It is also essential to consider combination therapy options to complement infused Treg, for instance low-dose interleukin-2 (IL-2) to support pre-existing and infused Treg survival and suppressive function. Understanding the hepatic microenvironment in both early- and late-stage AILD presents significant opportunity to better tailor Treg therapy in different patient groups. Modification of a hostile microenvironment to a more favourable one either prior to or during Treg therapy could enhance the efficacy and longevity of infused GMP-Treg. Applying recent technology to discovery of autoantigen responses in AILD, T cell receptor (TCR) sequencing and use of chimeric antigen receptor (CAR) technology represents the next frontier for disease-specific CAR-Treg therapies. Consideration of all these aspects in future trials and discovery research would position GMP Treg immunotherapy as a viable personalised-medicine treatment option for effective control of autoimmune liver diseases.
Collapse
|
11
|
Bai T, Yu S, Feng J. Advances in the Role of Endothelial Cells in Cerebral Small Vessel Disease. Front Neurol 2022; 13:861714. [PMID: 35481273 PMCID: PMC9035937 DOI: 10.3389/fneur.2022.861714] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/09/2022] [Indexed: 12/13/2022] Open
Abstract
Cerebral small vessel disease (CSVD) poses a serious socio-economic burden due to its high prevalence and severe impact on the quality of life of elderly patients. Pathological changes in CSVD mainly influence small cerebral arteries, microarteries, capillaries, and small veins, which are usually caused by multiple vascular risk factors. CSVD is often identified on brain magnetic resonance imaging (MRI) by recent small subcortical infarcts, white matter hyperintensities, lacune, cerebral microbleeds (CMBs), enlarged perivascular spaces (ePVSs), and brain atrophy. Endothelial cell (EC) dysfunction is earlier than clinical symptoms. Immune activation, inflammation, and oxidative stress may be potential mechanisms of EC injury. ECs of the blood–brain–barrier (BBB) are the most important part of the neurovascular unit (NVU) that ensures constant blood flow to the brain. Impaired cerebral vascular autoregulation and disrupted BBB cause cumulative brain damage. This review will focus on the role of EC injury in CSVD. Furthermore, several specific biomarkers will be discussed, which may be useful for us to assess the endothelial dysfunction and explore new therapeutic directions.
Collapse
|
12
|
Xu Q, Chen X, Yu T, Tang Q, Zhou Z, Wang H, Huang W, Huang T, Liang F. Downregulation of VAP-1 in OSCC suppresses tumor growth and metastasis via NF-κB/IL-8 signaling and reduces neutrophil infiltration. J Oral Pathol Med 2022; 51:332-341. [PMID: 35174543 DOI: 10.1111/jop.13285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 01/21/2022] [Accepted: 02/09/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Vascular adhesion protein-1 (VAP-1) is believed to play a role in inflammation. Studies have suggested that VAP-1-mediated activation of inflammation is dependent on NF-κB, leading to secretion of the interleukin(IL)-8; however, no reports have addressed the association between VAP-1 and NF-κB/IL-8 signaling in oral squamous cell carcinoma (OSCC). This study aimed to investigate the role of VAP-1 in OSCC and further explore whether VAP-1 is involved in the regulation of neutrophil infiltration in the tumor microenvironment (TME). METHODS Immunochemistry staining was used to observe VAP-1 expression. CCK-8 and Transwell assays were used to measure cell proliferation, migration, and invasion. OSCC xenograft mouse models were used for in vivo verification of the VAP-1 function. The expression of NF-κB and IL-8 were determined by qRT-PCR and western blot. ELISA for IL-8 was also conducted. The relationship between VAP-1 expression and neutrophil infiltration was analyzed by immunofluorescence. RESULTS VAP-1 was overexpressed in human OSCC tissues. Downregulation of VAP-1 suppressed OSCC cells proliferation, migration, and invasion in vitro and inhibited tumor proliferation and metastasis in vivo. Additionally, downregulation of VAP-1 inhibited NF-κB/IL-8 signaling in vitro and in vivo. VAP-1 expression was positively correlated with neutrophil infiltration in human OSCC tissues. Moreover, blocking VAP-1 decreased neutrophil infiltration by reducing IL-8 production. CONCLUSIONS VAP-1 downregulation in OSCC suppresses tumor growth and metastasis by inhibiting NF-κB/IL-8 signaling and reducing neutrophil infiltration in the TME, suggesting that VAP-1 may be a potential therapeutic target for OSCC.
Collapse
Affiliation(s)
- Qiongdong Xu
- Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, 530021, China.,Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Nanning, Guangxi, 530021, China.,Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, 530021, China
| | - Xueru Chen
- Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, 530021, China.,Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Nanning, Guangxi, 530021, China.,Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, 530021, China
| | - Tao Yu
- Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, 530021, China.,Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Nanning, Guangxi, 530021, China.,Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, 530021, China
| | - Qinchao Tang
- Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, 530021, China.,Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Nanning, Guangxi, 530021, China.,Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, 530021, China
| | - Zhuoqian Zhou
- Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, 530021, China.,Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Nanning, Guangxi, 530021, China.,Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, 530021, China
| | - Hongyu Wang
- Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, 530021, China.,Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Nanning, Guangxi, 530021, China.,Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, 530021, China
| | - Wanqian Huang
- Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Tianjing Huang
- Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Feixin Liang
- Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, 530021, China.,Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Nanning, Guangxi, 530021, China.,Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, 530021, China
| |
Collapse
|
13
|
Becchi S, Buson A, Balleine BW. Inhibition of vascular adhesion protein 1 protects dopamine neurons from the effects of acute inflammation and restores habit learning in the striatum. J Neuroinflammation 2021; 18:233. [PMID: 34654450 PMCID: PMC8520223 DOI: 10.1186/s12974-021-02288-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/04/2021] [Indexed: 12/17/2022] Open
Abstract
Background Changes in dopaminergic neural function can be induced by an acute inflammatory state that, by altering the integrity of the neurovasculature, induces neuronal stress, cell death and causes functional deficits. Effectively blocking these effects of inflammation could, therefore, reduce both neuronal and functional decline. To test this hypothesis, we inhibited vascular adhesion protein 1 (VAP-1), a membrane-bound protein expressed on the endothelial cell surface, that mediates leukocyte extravasation and induces oxidative stress. Method We induced dopaminergic neuronal loss by infusing lipopolysaccharide (LPS) directly into the substantia nigra (SN) in rats and administered the VAP-1 inhibitor, PXS-4681A, daily. Results LPS produced: an acute inflammatory response, the loss of dopaminergic neurons in the SN, reduced the dopaminergic projection to SN target regions, particularly the dorsolateral striatum (DLS), and a deficit in habit learning, a key function of the DLS. In an attempt to protect SN neurons from this inflammatory response we found that VAP-1 inhibition not only reduced neutrophil infiltration in the SN and striatum, but also reduced the associated striatal microglia and astrocyte response. We found VAP-1 inhibition protected dopamine neurons in the SN, their projections to the striatum and promoted the functional recovery of habit learning. Thus, we reversed the loss of habitual actions, a function usually dependent on dopamine release in DLS and sensitive to striatal dysfunction. Conclusions We establish, therefore, that VAP-1 inhibition has an anti-inflammatory profile that may be beneficial in the treatment of dopamine neuron dysfunction caused by an acute inflammatory state in the brain. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02288-8.
Collapse
Affiliation(s)
- Serena Becchi
- Decision Neuroscience Lab, School of Psychology, UNSW Sydney, Randwick, NSW, 2052, Australia
| | | | - Bernard W Balleine
- Decision Neuroscience Lab, School of Psychology, UNSW Sydney, Randwick, NSW, 2052, Australia.
| |
Collapse
|
14
|
Ibrahim SH. Sinusoidal endotheliopathy in nonalcoholic steatohepatitis: therapeutic implications. Am J Physiol Gastrointest Liver Physiol 2021; 321:G67-G74. [PMID: 34037463 PMCID: PMC8321796 DOI: 10.1152/ajpgi.00009.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Liver sinusoidal endothelial cells (LSECs) are distinct subtypes of endothelial cells lining a low flow vascular bed at the interface of the liver parenchyma and the circulating immune cells and soluble factors. Emerging literature implicates LSEC in the pathogenesis and progression of nonalcoholic fatty liver disease (NAFLD). During the evolution of NAFLD, LSEC dysfunction ensues. LSECs undergo morphological and functional transformation known as "capillarization," as well as a pathogenic increase in surface adhesion molecules expression, referred to in this review as "endotheliopathy." LSECs govern the composition of hepatic immune cell populations in nonalcoholic steatohepatis (NASH) by mediating leukocyte subset adhesion through specific combinations of activated adhesion molecules and secreted chemokines. Moreover, extracellular vesicles released by hepatocyte under lipotoxic stress in NASH act as a catalyst for the inflammatory response and promote immune cell chemotaxis and adhesion. In the current review, we highlight leukocyte adhesion to LSEC as an initiating event in the sterile inflammatory response in NASH. We discuss preclinical studies targeting immune cells adhesion in NASH mouse models and potential therapeutic anti-inflammatory strategies for human NASH.
Collapse
Affiliation(s)
- Samar H. Ibrahim
- 1Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota,2Division of Pediatric Gastroenterology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
15
|
Unzeta M, Hernàndez-Guillamon M, Sun P, Solé M. SSAO/VAP-1 in Cerebrovascular Disorders: A Potential Therapeutic Target for Stroke and Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22073365. [PMID: 33805974 PMCID: PMC8036996 DOI: 10.3390/ijms22073365] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/24/2022] Open
Abstract
The semicarbazide-sensitive amine oxidase (SSAO), also known as vascular adhesion protein-1 (VAP-1) or primary amine oxidase (PrAO), is a deaminating enzyme highly expressed in vessels that generates harmful products as a result of its enzymatic activity. As a multifunctional enzyme, it is also involved in inflammation through its ability to bind and promote the transmigration of circulating leukocytes into inflamed tissues. Inflammation is present in different systemic and cerebral diseases, including stroke and Alzheimer’s disease (AD). These pathologies show important affectations on cerebral vessels, together with increased SSAO levels. This review summarizes the main roles of SSAO/VAP-1 in human physiology and pathophysiology and discusses the mechanisms by which it can affect the onset and progression of both stroke and AD. As there is an evident interrelationship between stroke and AD, basically through the vascular system dysfunction, the possibility that SSAO/VAP-1 could be involved in the transition between these two pathologies is suggested. Hence, its inhibition is proposed to be an interesting therapeutical approach to the brain damage induced in these both cerebral pathologies.
Collapse
Affiliation(s)
- Mercedes Unzeta
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences, Universitat Auònoma de Barcelona, 08193 Barcelona, Spain;
| | - Mar Hernàndez-Guillamon
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
- Correspondence: ; Tel.: +34-934-896-766
| | - Ping Sun
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Montse Solé
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
| |
Collapse
|
16
|
Shepherd EL, Karim S, Newsome PN, Lalor PF. Inhibition of vascular adhesion protein-1 modifies hepatic steatosis in vitro and in vivo. World J Hepatol 2020; 12:931-948. [PMID: 33312420 PMCID: PMC7701969 DOI: 10.4254/wjh.v12.i11.931] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/23/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is associated with obesity, insulin resistance and dyslipidaemia and currently is estimated to affect up to a third of all individuals in developed countries. Current standard of care for patients varies according to disease stage, but includes lifestyle interventions common insulin sensitizers, antioxidants and lipid modifiers. However, to date specific therapies have shown little histological or fibrosis stage improvement in large clinical trials, and there is still no licensed therapy for NAFLD. Given the high prevalence, limited treatment options and significant screening costs for the general population, new treatments are urgently required.
AIM To assess the potential for inhibition of the amine oxidase enzyme vascular adhesion protein-1 (VAP-1) to modify hepatic lipid accumulation in NAFLD.
METHODS We have used immunochemical and qPCR analysis to document expression of VAP-1 and key functional proteins and transporters across the NAFLD spectrum. We then utilised hepatocytes in culture and human precision cut liver slices in concert with selective enzyme activity inhibitors to test the effects of activating the semicarbazide-sensitive amine oxidase activity of VAP-1 on hepatic lipid uptake and triglyceride export. A murine model of NAFLD was also used to determine the consequences of VAP-1 knockout and gene expression arrays were used to quantify the effects of VAP-1 activity on key lipid modifying and proinflammatory gene expression.
RESULTS We confirmed that increasing severity of NAFLD and progression to cirrhosis was associated with a significant increase in hepatocellular VAP-1 expression. Hepatocytes in vitro exposed to recombinant VAP-1 and its substrate methylamine showed increased lipid accumulation as determined by quantification of Oil Red O uptake. This was recapitulated using hydrogen peroxide, and lipid accumulation was accompanied by changes in expression of the lipid transporter molecules FABP3, FATP6, insulin receptor subunits and PPARα. Human liver tissue exposed to recombinant VAP-1 or substrates for endo/exogenous VAP-1 produced less triglyceride than untreated tissue and demonstrated an increase in steatosis. This response could be inhibited by using bromoethylamine to inhibit the SSAO activity of VAP-1, and mice deficient in VAP-1/AOC3 also demonstrated reduced steatosis on high fat diet. Exposure of human liver tissue to methylamine to activate VAP-1 resulted in increased expression of FABP2 and 4, FATP3-5, caveolin-1, VLDLR, PPARGC1 and genes associated with the inflammatory response.
CONCLUSION Our data confirm that the elevations in hepatic VAP-1 expression reported in nonalcoholic steatohepatitis can contribute to steatosis, metabolic disturbance and inflammation. This suggests that targeting the semicarbazide sensitive amine oxidase capacity of VAP-1 may represent a useful adjunct to other therapeutic strategies in NAFLD.
Collapse
Affiliation(s)
- Emma L Shepherd
- Centre for Liver and Gastroenterology Research, Birmingham National Institute for Health Research, Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, West Midlands, United Kingdom
| | - Sumera Karim
- Centre for Liver and Gastroenterology Research, Birmingham National Institute for Health Research, Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, West Midlands, United Kingdom
| | - Philip N Newsome
- Centre for Liver and Gastroenterology Research, Birmingham National Institute for Health Research, Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, West Midlands, United Kingdom
- Liver Unit, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham B15 2TT, West Midlands, United Kingdom
| | - Patricia F Lalor
- Centre for Liver and Gastroenterology Research, Birmingham National Institute for Health Research, Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, West Midlands, United Kingdom
| |
Collapse
|
17
|
Emerging Roles of Liver Sinusoidal Endothelial Cells in Nonalcoholic Steatohepatitis. BIOLOGY 2020; 9:biology9110395. [PMID: 33198153 PMCID: PMC7697091 DOI: 10.3390/biology9110395] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022]
Abstract
Simple Summary Nonalcoholic fatty liver disease (NAFLD) is a hepatic manifestation of the metabolic syndrome. With the prevalence of obesity and type 2 diabetes, NAFLD is becoming the most common liver disorder worldwide. More than 10% of NAFLD patients progress to an inflammatory and fibrotic form called nonalcoholic steatohepatitis (NASH), which can lead to end-stage liver disease. Liver sinusoidal endothelial cells (LSEC) are highly specialized cells located at the interface between the flowing blood in the liver and the other liver cells. The current review highlights the recent knowledge of the role of LSEC in the development of NASH, and how LSEC change their structure and function during NAFLD progression. Moreover, the review discusses the pathogenic role of nanometer-sized particles called extracellular vesicles that mediate intercellular communication in the NASH liver. The current manuscript has a special emphasis on the role of adhesion molecules expressed on the LSEC surface in the recruitment of circulating leukocytes to the liver, a critical step in liver inflammation in NASH. Furthermore, the review shed some lights on LSEC-targeted potential therapeutic strategies in NASH. Abstract Nonalcoholic steatohepatitis (NASH) has become a growing public health problem worldwide, yet its pathophysiology remains unclear. Liver sinusoidal endothelial cells (LSEC) have unique morphology and function, and play a critical role in liver homeostasis. Emerging literature implicates LSEC in many pathological processes in the liver, including metabolic dysregulation, inflammation, angiogenesis, and carcinogenesis. In this review, we highlight the current knowledge of the role of LSEC in each of the progressive phases of NASH pathophysiology (steatosis, inflammation, fibrosis, and the development of hepatocellular carcinoma). We discuss processes that have important roles in NASH progression including the detrimental transformation of LSEC called “capillarization”, production of inflammatory and profibrogenic mediators by LSEC as well as LSEC-mediated angiogenesis. The current review has a special emphasis on LSEC adhesion molecules, and their key role in the inflammatory response in NASH. Moreover, we discuss the pathogenic role of extracellular vesicles and their bioactive cargos in liver intercellular communication, inflammation, and fibrosis. Finally, we highlight LSEC-adhesion molecules and derived bioactive product as potential therapeutic targets for human NASH.
Collapse
|
18
|
Öksüz Z, Üçbilek E, Serin MS, Yaraş S, Temel GO, Sezgin O. Circulating vascular adhesion protein-1(VAP-1): a possible biomarker for liver fibrosis associated with chronic hepatitis B and C. Braz J Microbiol 2020; 51:1757-1763. [PMID: 32959205 DOI: 10.1007/s42770-020-00379-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/09/2020] [Indexed: 12/17/2022] Open
Abstract
Vascular adhesion protein-1 (VAP-1) is a multifunctional protein that plays a role in chronic liver diseases and fibrogenesis. The present study aimed to investigate the possible association of VAP-1 levels with the severity of disease progression in chronic hepatitis (CH) B and C patients with differing stages of fibrosis (F0-4), CHB/CHC-related cirrhosis, and hepatocellular carcinoma (HCC). The VAP-1 concentration in patient sera was determined by ELISA. The VAP-1 levels were compared between the F0 group and the F1, F2, F3, F4, cirrhosis, and HCC groups of CHB patients and between the F1 group and the F2, F3, F4, cirrhosis, and HCC groups of CHC patients. The levels of VAP-1 were significantly increased in CHB patients with progressive stages of fibrosis, with the highest concentration being found in those with stage F4 (severe fibrosis). A statistically significant difference was found between F0 and F4 in patients with CHB, but no statistically significant difference was observed between F1 and F4 in patients with CHC. Interestingly, there was no statistically significant difference in VAP-1 levels between patients with cirrhosis and HCC (either CHB or CHC, independently). Moreover, no relationship was found between VAP-1 and ALT levels in either CHC or CHB patients. In general, the VAP-1 levels were significantly higher in CHB than in CHC patients (P < 0.01). In conclusion, we suggest that the VAP-1 level may be a noninvasive biomarker for monitoring the severity of fibrogenesis in patients with hepatitis B infection.
Collapse
Affiliation(s)
- Zehra Öksüz
- Department of Pharmaceutical Microbiology, Mersin University Faculty of Pharmacy, Mersin, Turkey.
| | - Enver Üçbilek
- Department of Gastroenterology, Mersin University Faculty of Medicine, Mersin, Turkey
| | - Mehmet Sami Serin
- Department of Pharmaceutical Microbiology, Mersin University Faculty of Pharmacy, Mersin, Turkey
| | - Serkan Yaraş
- Department of Gastroenterology, Mersin University Faculty of Medicine, Mersin, Turkey
| | - Gülhan Orekici Temel
- Department of Biostatistics, Mersin University Faculty of Medicine, Mersin, Turkey
| | - Orhan Sezgin
- Department of Gastroenterology, Mersin University Faculty of Medicine, Mersin, Turkey
| |
Collapse
|
19
|
Wilkinson AL, Qurashi M, Shetty S. The Role of Sinusoidal Endothelial Cells in the Axis of Inflammation and Cancer Within the Liver. Front Physiol 2020; 11:990. [PMID: 32982772 PMCID: PMC7485256 DOI: 10.3389/fphys.2020.00990] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
Liver sinusoidal endothelial cells (LSEC) form a unique barrier between the liver sinusoids and the underlying parenchyma, and thus play a crucial role in maintaining metabolic and immune homeostasis, as well as actively contributing to disease pathophysiology. Whilst their endocytic and scavenging function is integral for nutrient exchange and clearance of waste products, their capillarisation and dysfunction precedes fibrogenesis. Furthermore, their ability to promote immune tolerance and recruit distinct immunosuppressive leukocyte subsets can allow persistence of chronic viral infections and facilitate tumour development. In this review, we present the immunological and barrier functions of LSEC, along with their role in orchestrating fibrotic processes which precede tumourigenesis. We also summarise the role of LSEC in modulating the tumour microenvironment, and promoting development of a pre-metastatic niche, which can drive formation of secondary liver tumours. Finally, we summarise closely inter-linked disease pathways which collectively perpetuate pathogenesis, highlighting LSEC as novel targets for therapeutic intervention.
Collapse
Affiliation(s)
| | | | - Shishir Shetty
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
20
|
Abstract
The human liver is an organ with a diverse array of immunologic functions. Its unique anatomic position that leads to it receiving all the mesenteric venous blood, combined with its unique micro anatomy, allows it to serve as a sentinel for the body's immune system. Hepatocytes, biliary epithelial cells, Kupffer cells, stellate cells, and liver sinusoidal endothelial cells express key molecules that recruit and activate innate and adaptive immunity. Additionally, a diverse array of lymphoid and myeloid immune cells resides within and traffics to the liver in specific circumstances. Derangement of these trafficking mechanisms underlies the pathophysiology of autoimmune liver diseases, nonalcoholic steatohepatitis, and liver transplantation. Here, we review these pathways and interactions along with potential targets that have been identified to be exploited for therapeutic purposes.
Collapse
|
21
|
Sun X, Harris EN. New aspects of hepatic endothelial cells in physiology and nonalcoholic fatty liver disease. Am J Physiol Cell Physiol 2020; 318:C1200-C1213. [PMID: 32374676 DOI: 10.1152/ajpcell.00062.2020] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The liver is the central metabolic hub for carbohydrate, lipid, and protein metabolism. It is composed of four major types of cells, including hepatocytes, endothelial cells (ECs), Kupffer cells, and stellate cells. Hepatic ECs are highly heterogeneous in both mice and humans, representing the second largest population of cells in liver. The majority of them line hepatic sinusoids known as liver sinusoidal ECs (LSECs). The structure and biology of LSECs and their roles in physiology and liver disease were reviewed recently. Here, we do not give a comprehensive review of LSEC structure, function, or pathophysiology. Instead, we focus on the recent progress in LSEC research and other hepatic ECs in physiology and nonalcoholic fatty liver disease and other hepatic fibrosis-related conditions. We discuss several current areas of interest, including capillarization, scavenger function, autophagy, cellular senescence, paracrine effects, and mechanotransduction. In addition, we summarize the strengths and weaknesses of evidence for the potential role of endothelial-to-mesenchymal transition in liver fibrosis.
Collapse
Affiliation(s)
- Xinghui Sun
- Department of Biochemistry, University of Nebraska-Lincoln, Beadle Center, Lincoln, Nebraska.,Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Nebraska.,Nebraska Center for the Prevention of Obesity Diseases through Dietary Molecules, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Edward N Harris
- Department of Biochemistry, University of Nebraska-Lincoln, Beadle Center, Lincoln, Nebraska.,Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Nebraska.,Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
22
|
Méndez-Sánchez N, Valencia-Rodríguez A, Coronel-Castillo C, Vera-Barajas A, Contreras-Carmona J, Ponciano-Rodríguez G, Zamora-Valdés D. The cellular pathways of liver fibrosis in non-alcoholic steatohepatitis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:400. [PMID: 32355844 PMCID: PMC7186641 DOI: 10.21037/atm.2020.02.184] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 02/29/2020] [Indexed: 12/12/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) is considered the advanced stage of non-alcoholic fatty liver disease (NAFLD). It is characterized by liver steatosis, inflammation and different degrees of fibrosis. Although the exact mechanisms by which fatty liver progresses to NASH are still not well understood, innate and adaptive immune responses seem to be essential key regulators in the establishment, progression, and chronicity of these disease. Diet-induced lipid overload of parenchymal and non-parenchymal liver cells is considered the first step for the development of fatty liver with the consequent organelle dysfunction, cellular stress and liver injury. These will generate the production of pro-inflammatory cytokines, chemokines and damage-associated molecular patterns (DAMPs) that will upregulate the activation of Kupffer cells (KCs) and monocyte-derived macrophages (MMs) favoring the polarization of the tolerogenic environment of the liver to an immunogenic phenotype with the resulting transdifferentiation of hepatic stellate cells (HSCs) into myofibroblasts developing fibrosis. In the long run, dendritic cells (DCs) will activate CD4+ T cells polarizing into the pro-inflammatory lymphocytes Th1 and Th17 worsening the liver damage and inflammation. Therefore, the objective of this review is to discuss in a systematic way the mechanisms known so far of the immune and non-proper immune liver cells in the development and progression of NASH.
Collapse
Affiliation(s)
- Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
- Faculty of Medicine. National Autonomous University of Mexico, Mexico City, Mexico
| | | | | | | | | | | | | |
Collapse
|
23
|
Yordanov Y. Caffeic acid phenethyl ester (CAPE): pharmacodynamics and potential for therapeutic application. PHARMACIA 2019. [DOI: 10.3897/pharmacia.66.e38573] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Caffeic acid phenethyl ester (CAPE) is the major pharmacologically-active component of some propolis types, rich in polyphenols, such as poplar propolis types. CAPE has the potential to be applied as a pharmaceutical as it possesses most of the pharmacological activities of propolis, such as anti-proliferative, antioxidant, immunomodulatory, antidiabetic, anti-inflammatory and antimicrobial. Its advantage is that it lacks some of the downsides of total propolis extracts, such as inability for unified standardization, which is cornerstone for implementing its therapeutic potential as a drug. The current paper provides an overview on the pharmacodynamic principles of CAPE. We present literature search outcomes form ClinicalTrials.gov database and from scientific publications, available on Scopus and Crossref databases. We take a round view of CAPE’s potential therapeutic implications in light of approved drugs with related modes of action.
Collapse
|
24
|
de Krijger M, Wildenberg ME, de Jonge WJ, Ponsioen CY. Return to sender: Lymphocyte trafficking mechanisms as contributors to primary sclerosing cholangitis. J Hepatol 2019; 71:603-615. [PMID: 31108158 DOI: 10.1016/j.jhep.2019.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/29/2019] [Accepted: 05/09/2019] [Indexed: 12/14/2022]
Abstract
Primary sclerosing cholangitis (PSC) is an inflammatory disease of the biliary tree, characterised by stricturing bile duct disease and progression to liver fibrosis. The pathophysiology of PSC is still unknown. The concurrence with inflammatory bowel disease (IBD) in about 70% of cases has led to the hypothesis that gut-homing lymphocytes aberrantly traffic to the liver, contributing to disease pathogenesis in patients with both PSC and IBD (PSC-IBD). The discovery of mutual trafficking pathways of lymphocytes to target tissues, and expression of gut-specific adhesion molecules and chemokines in the liver has pointed in this direction. There is now increasing interest in using drugs that intervene with these trafficking pathways (e.g. vedolizumab, etrolizumab) for the treatment of PSC-IBD. In this review we discuss what is currently known about the immunological interactions between the gut and the liver in concomitant PSC and IBD, as well as potential therapeutic options for intervening in these mechanisms.
Collapse
Affiliation(s)
- Manon de Krijger
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Manon E Wildenberg
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Wouter J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Surgery, University of Bonn, Bonn, Germany
| | - Cyriel Y Ponsioen
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
25
|
Solé M, Esteban-Lopez M, Taltavull B, Fábregas C, Fadó R, Casals N, Rodríguez-Álvarez J, Miñano-Molina AJ, Unzeta M. Blood-brain barrier dysfunction underlying Alzheimer's disease is induced by an SSAO/VAP-1-dependent cerebrovascular activation with enhanced Aβ deposition. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2189-2202. [PMID: 31047972 DOI: 10.1016/j.bbadis.2019.04.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/03/2019] [Accepted: 04/26/2019] [Indexed: 12/11/2022]
Abstract
Dysfunctions of the vascular system directly contribute to the onset and progression of Alzheimer's disease (AD). The blood-brain barrier (BBB) shows signs of malfunction at early stages of the disease. When Abeta peptide (Aβ) is deposited on brain vessels, it induces vascular degeneration by producing reactive oxygen species and promoting inflammation. These molecular processes are also related to an excessive SSAO/VAP-1 (semicarbazide-sensitive amine oxidase) enzymatic activity, observed in plasma and in cerebrovascular tissue of AD patients. We studied the contribution of vascular SSAO/VAP-1 to the BBB dysfunction in AD using in vitro BBB models. Our results show that SSAO/VAP-1 expression is associated to endothelial activation by altering the release of pro-inflammatory and pro-angiogenic angioneurins, most highly IL-6, IL-8 and VEGF. It is also related to a BBB structure alteration, with a decrease in tight-junction proteins such as zona occludens or claudin-5. Moreover, the BBB function reveals increased permeability and leukocyte adhesion in cells expressing SSAO/VAP-1, as well as an enhancement of the vascular Aβ deposition induced by mechanisms both dependent and independent of the enzymatic activity of SSAO/VAP-1. These results reveal an interesting role of vascular SSAO/VAP-1 in BBB dysfunction related to AD progression, opening a new window in the search of alternative therapeutic targets for fighting AD.
Collapse
Affiliation(s)
- Montse Solé
- Biochemistry and Molecular Biology Department, Institute of Neurosciences (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain.
| | - María Esteban-Lopez
- Biochemistry and Molecular Biology Department, Institute of Neurosciences (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain
| | - Biel Taltavull
- Biochemistry and Molecular Biology Department, Institute of Neurosciences (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain
| | - Cristina Fábregas
- Biochemistry and Molecular Biology Department, Institute of Neurosciences (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain
| | - Rut Fadó
- Basic Sciences Department, Facultat de Medicina i Ciències de la Salut, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Núria Casals
- Basic Sciences Department, Facultat de Medicina i Ciències de la Salut, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Santiago de Compostela, Spain
| | - Jose Rodríguez-Álvarez
- Biochemistry and Molecular Biology Department, Institute of Neurosciences (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Alfredo J Miñano-Molina
- Biochemistry and Molecular Biology Department, Institute of Neurosciences (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Mercedes Unzeta
- Biochemistry and Molecular Biology Department, Institute of Neurosciences (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain.
| |
Collapse
|
26
|
Abstract
Significance: Vascular adhesion protein-1 (VAP-1) is an ectoenzyme that oxidates primary amines in a reaction producing also hydrogen peroxide. VAP-1 on the blood vessel endothelium regulates leukocyte extravasation from the blood into tissues under physiological and pathological conditions. Recent Advances: Inhibition of VAP-1 by neutralizing antibodies and by several novel small-molecule enzyme inhibitors interferes with leukocyte trafficking and alleviates inflammation in many experimental models. Targeting of VAP-1 also shows beneficial effects in several other diseases, such as ischemia/reperfusion, fibrosis, and cancer. Moreover, soluble VAP-1 levels may serve as a new prognostic biomarker in selected diseases. Critical Issues: Understanding the contribution of the enzyme activity-independent and enzyme activity-dependent functions, which often appear to be mediated by the hydrogen peroxide production, in the VAP-1 biology will be crucial. Similarly, there is a pressing need to understand which of the VAP-1 functions are regulated through the modulation of leukocyte trafficking, and what is the role of VAP-1 synthesized in adipose and smooth muscle cells. Future Directions: The specificity and selectivity of new VAP-1 inhibitors, and their value in animal models under therapeutic settings need to be addressed. Results from several programs studying the therapeutic potential of VAP-1 inhibition, which now are in clinical trials, will reveal the relevance of this amine oxidase in humans.
Collapse
Affiliation(s)
- Marko Salmi
- 1 MediCity , Turku, Finland .,2 Institute of Biomedicine, University of Turku, Turku, Finland
| | - Sirpa Jalkanen
- 1 MediCity , Turku, Finland .,2 Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
27
|
Increased Circulating VAP-1 Levels Are Associated with Liver Fibrosis in Chronic Hepatitis C Infection. J Clin Med 2019; 8:jcm8010103. [PMID: 30658395 PMCID: PMC6352124 DOI: 10.3390/jcm8010103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 12/12/2022] Open
Abstract
Vascular adhesion protein-1 (VAP-1) is a multifunction protein. While membrane-bound VAP-1 is an adhesion protein, soluble VAP-1 catalyzes the deamination of primary amines through its semicarbazide-sensitive amino oxidase (SSAO) activity. VAP-1 supports the transmigration of leukocytes and increases oxidative stress. In chronic liver diseases, it plays a role in leukocyte infiltration and fibrogenesis. Here, we measured VAP-1 plasma concentration and its SSAO activity in 322 patients with chronic hepatitis C infection and evaluated the association of VAP-1 with fibrosis stages. VAP-1 concentration strongly correlated with liver stiffness and was the second strongest influencing variable after gamma-glutamytransferase (GGT) for liver stiffness in regression analysis. The VAP-1 concentration increased with advancing fibrosis stages and the highest concentrations were found in patients with cirrhosis. According to the receiver operating characteristic (ROC) analysis, a VAP-1 cut-off value of 541 ng/mL predicted histologically confirmed cirrhosis (sensitivity 74%; specificity 72%). SSAO activity correlated only moderately with liver stiffness, showing a relatively small increase in advanced fibrosis. To our knowledge, this is the first study on VAP-1 in chronic hepatitis C infection showing its association with progressive fibrosis. In conclusion, VAP-1 plasma concentration, rather than its SSAO activity, may represent a non-invasive biomarker for monitoring fibrogenesis in patients with chronic hepatitis C infection.
Collapse
|
28
|
Kim L, Lim Y, Park SY, Kim YJ, Kwon O, Lee JH, Shin JH, Yang YK, Kim JY. A comparative study of the antithrombotic effect through activated endothelium of garlic powder and tomato extracts using a rodent model of collagen and epinephrine induced thrombosis. Food Sci Biotechnol 2018; 27:1513-1518. [PMID: 30319862 DOI: 10.1007/s10068-018-0469-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/25/2018] [Accepted: 09/04/2018] [Indexed: 10/28/2022] Open
Abstract
In this study, garlic powder, tomato extract and a mixture of both were analyzed for anti-thrombotic effects using a collagen and epinephrine induced thrombosis model. Rats were randomly assigned to control, thrombosis induced control (COL/EP), garlic powder (G), tomato extract (T) and mixture of garlic powder and tomato extract (GT) groups. Test materials were administered for 7 days and thrombosis was induced by collagen and epinephrine injection. The results showed that G, T, and GT delayed activated partial thromboplastin time and reduced the expression of intracellular adhesion molecule-1 mRNA. Histological analysis of aorta and lung revealed that thrombosis was partially improved by G, T, and GT. Although there was no synergistic effect in GT compared to G and T treatment, this study showed that G, T, and GT have anti-thrombotic effect.
Collapse
Affiliation(s)
- Leeseon Kim
- 1Department of Food Science and Technology, Seoul National University of Science and Technology, Seoul, 01811 Republic of Korea
| | - Yeni Lim
- 2Department of Nutritional Science and Food Management, Ewha Woman's University, Seoul, 03760 Republic of Korea
| | - Soo-Yeon Park
- 2Department of Nutritional Science and Food Management, Ewha Woman's University, Seoul, 03760 Republic of Korea
| | - You Jin Kim
- 2Department of Nutritional Science and Food Management, Ewha Woman's University, Seoul, 03760 Republic of Korea
| | - Oran Kwon
- 2Department of Nutritional Science and Food Management, Ewha Woman's University, Seoul, 03760 Republic of Korea
| | - Jin Hee Lee
- 3Department of Food Science and Biotechnology, CHA University, Seongnam, Gyeonggido 13488 Republic of Korea
| | - Jae-Ho Shin
- 4Department of Biomedical Laboratory Science, Eulji University, Seongnam, Gyeonggido 13135 Republic of Korea
| | - Yoon Kyoung Yang
- 5Department of Nutritional Science and Food Management, Soongeui Women's College, Seoul, 04628 Republic of Korea
| | - Ji Yeon Kim
- 1Department of Food Science and Technology, Seoul National University of Science and Technology, Seoul, 01811 Republic of Korea
| |
Collapse
|
29
|
Trivedi PJ, Tickle J, Vesterhus MN, Eddowes PJ, Bruns T, Vainio J, Parker R, Smith D, Liaskou E, Thorbjørnsen LW, Hirschfield GM, Auvinen K, Hubscher SG, Salmi M, Adams DH, Weston CJ. Vascular adhesion protein-1 is elevated in primary sclerosing cholangitis, is predictive of clinical outcome and facilitates recruitment of gut-tropic lymphocytes to liver in a substrate-dependent manner. Gut 2018; 67:1135-1145. [PMID: 28428344 PMCID: PMC5969351 DOI: 10.1136/gutjnl-2016-312354] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 03/23/2017] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Primary sclerosing cholangitis (PSC) is the classical hepatobiliary manifestation of IBD. This clinical association is linked pathologically to the recruitment of mucosal T cells to the liver, via vascular adhesion protein (VAP)-1-dependent enzyme activity. Our aim was to examine the expression, function and enzymatic activation of the ectoenzyme VAP-1 in patients with PSC. DESIGN We examined VAP-1 expression in patients with PSC, correlated levels with clinical characteristics and determined the functional consequences of enzyme activation by specific enzyme substrates on hepatic endothelium. RESULTS The intrahepatic enzyme activity of VAP-1 was elevated in PSC versus immune-mediated disease controls and non-diseased liver (p<0.001). The adhesion of gut-tropic α4β7+lymphocytes to hepatic endothelial cells in vitro under flow was attenuated by 50% following administration of the VAP-1 inhibitor semicarbazide (p<0.01). Of a number of natural VAP-1 substrates tested, cysteamine-which can be secreted by inflamed colonic epithelium and gut bacteria-was the most efficient (yielded the highest enzymatic rate) and efficacious in its ability to induce expression of functional mucosal addressin cell adhesion molecule-1 on hepatic endothelium. In a prospectively evaluated patient cohort with PSC, elevated serum soluble (s)VAP-1 levels predicted poorer transplant-free survival for patients, independently (HR: 3.85, p=0.003) and additively (HR: 2.02, p=0.012) of the presence of liver cirrhosis. CONCLUSIONS VAP-1 expression is increased in PSC, facilitates adhesion of gut-tropic lymphocytes to liver endothelium in a substrate-dependent manner, and elevated levels of its circulating form predict clinical outcome in patients.
Collapse
Affiliation(s)
- Palak J Trivedi
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK,Liver Unit, University Hospitals Birmingham Queen Elizabeth, Birmingham, UK
| | - Joseph Tickle
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Mette Nåmdal Vesterhus
- Department of Transplantation Medicine, Division of Cancer Medicine, Surgery and Transplantation, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway,National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway
| | - Peter J Eddowes
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Tony Bruns
- Department of Internal Medicine IV, University Hospital Jena, Germany,Center for Sepsis Control and Care, University Hospital Jena, Germany
| | | | - Richard Parker
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK,Liver Unit, University Hospitals Birmingham Queen Elizabeth, Birmingham, UK
| | | | - Evaggelia Liaskou
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Liv Wenche Thorbjørnsen
- Department of Transplantation Medicine, Division of Cancer Medicine, Surgery and Transplantation, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway,National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway
| | - Gideon M Hirschfield
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK,Liver Unit, University Hospitals Birmingham Queen Elizabeth, Birmingham, UK
| | - Kaisa Auvinen
- MediCity Research Laboratory, University of Turku, Turku, Finland,Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - Stefan G Hubscher
- Department of Cellular Pathology, University Hospitals Birmingham Queen Elizabeth, Birmingham, UK
| | - Marko Salmi
- MediCity Research Laboratory, University of Turku, Turku, Finland,Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - David H Adams
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK,Liver Unit, University Hospitals Birmingham Queen Elizabeth, Birmingham, UK
| | - Chris J Weston
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| |
Collapse
|
30
|
Mehrfeld C, Zenner S, Kornek M, Lukacs-Kornek V. The Contribution of Non-Professional Antigen-Presenting Cells to Immunity and Tolerance in the Liver. Front Immunol 2018; 9:635. [PMID: 29643856 PMCID: PMC5882789 DOI: 10.3389/fimmu.2018.00635] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/14/2018] [Indexed: 12/12/2022] Open
Abstract
The liver represents a unique organ biased toward a tolerogenic milieu. Due to its anatomical location, it is constantly exposed to microbial and food-derived antigens from the gut and thus equipped with a complex cellular network that ensures dampening T-cell responses. Within this cellular network, parenchymal cells (hepatocytes), non-parenchymal cells (liver sinusoidal endothelial cells and hepatic stellate cells), and immune cells contribute directly or indirectly to this process. Despite this refractory bias, the liver is capable of mounting efficient T-cell responses. How the various antigen-presenting cell (APC) populations contribute to this process and how they handle danger signals determine the outcome of the generated immune responses. Importantly, liver mounted responses convey consequences not only for the local but also to systemic immunity. Here, we discuss various aspects of antigen presentation and its consequences by the non-professional APCs in the liver microenvironment.
Collapse
Affiliation(s)
- Christina Mehrfeld
- Department of Medicine II, Saarland University Medical Center, Homburg, Germany
| | - Steven Zenner
- Department of Medicine II, Saarland University Medical Center, Homburg, Germany
| | - Miroslaw Kornek
- Department of Medicine II, Saarland University Medical Center, Homburg, Germany
| | | |
Collapse
|
31
|
Abstract
Liver sinusoidal endothelial cells (LSECs) line the low shear, sinusoidal capillary channels of the liver and are the most abundant non-parenchymal hepatic cell population. LSECs do not simply form a barrier within the hepatic sinusoids but have vital physiological and immunological functions, including filtration, endocytosis, antigen presentation and leukocyte recruitment. Reflecting these multifunctional properties, LSECs display unique structural and phenotypic features that differentiate them from the capillary endothelium present within other organs. It is now clear that LSECs have a critical role in maintaining immune homeostasis within the liver and in mediating the immune response during acute and chronic liver injury. In this Review, we outline how LSECs influence the immune microenvironment within the liver and discuss their contribution to immune-mediated liver diseases and the complications of fibrosis and carcinogenesis.
Collapse
|
32
|
Sun P, Hernandez-Guillamón M, Campos-Martorell M, Simats A, Montaner J, Unzeta M, Solé M. Simvastatin blocks soluble SSAO/VAP-1 release in experimental models of cerebral ischemia: Possible benefits for stroke-induced inflammation control. Biochim Biophys Acta Mol Basis Dis 2017; 1864:542-553. [PMID: 29175057 DOI: 10.1016/j.bbadis.2017.11.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/19/2017] [Accepted: 11/20/2017] [Indexed: 12/11/2022]
Abstract
Beyond cholesterol reduction, statins mediate their beneficial effects on stroke patients through pleiotropic actions. They have shown anti-inflammatory properties by a number of different mechanisms, including the inhibition of NF-κB transcriptional activity and the consequent increase and release of adhesion molecules. We have studied simvastatin's effects on the vascular enzyme semicarbazide-sensitive amine oxidase/vascular adhesion protein 1 (SSAO/VAP-1), which is involved in stroke-mediated brain injury. SSAO/VAP-1 has leukocyte-binding capacity and mediates the expression of other adhesion proteins through signaling molecules generated by its catalytic activity. Our results indicate that soluble SSAO/VAP-1 is released into the bloodstream after an ischemic stimulus, in parallel with an increase in E-selectin and VCAM-1 and correlating with infarct volume. Simvastatin blocks soluble SSAO/VAP-1 release and prevents E-selectin and VCAM-1 overexpression as well. Simvastatin also effectively blocks SSAO/VAP-1-mediated leukocyte adhesion, although it is not an enzymatic inhibitor of SSAO in vitro. In addition, simvastatin-induced changes in adhesion molecules are greater in human brain endothelial cell cultures expressing SSAO/VAP-1, compared to those not expressing it, indicating some synergic effect with SSAO/VAP-1. We think that part of the beneficial effect of simvastatin in stroke is mediated by the attenuation of the SSAO/VAP-1-dependent inflammatory response.
Collapse
Affiliation(s)
- Ping Sun
- Biochemistry and Molecular Biology Department, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Mar Hernandez-Guillamón
- Neurovascular Research Laboratory, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mireia Campos-Martorell
- Neurovascular Research Laboratory, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alba Simats
- Neurovascular Research Laboratory, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mercedes Unzeta
- Biochemistry and Molecular Biology Department, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.
| | - Montse Solé
- Biochemistry and Molecular Biology Department, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.
| |
Collapse
|
33
|
Korošec T, Tomažin U, Horvat S, Keber R, Salobir J. The diverse effects of α- and γ-tocopherol on chicken liver transcriptome. Poult Sci 2017; 96:667-680. [PMID: 27587731 DOI: 10.3382/ps/pew296] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 07/11/2016] [Indexed: 01/11/2023] Open
Abstract
α-Tocopherol is the form of vitamin E with the highest biological value and is almost exclusively considered as vitamin E in feed and feed supplements. Because γ-tocopherol, the predominant form of vitamin E naturally present in chicken feed, is not considered as a source of vitamin E, its re-evaluation with newer methods might be important.Despite γ-tocopherol's lower estimated biological value, it has been shown to be effective in reducing reactive nitrogen species, regulating immune and inflammatory processes, and diminishing the risk of metabolic perturbations and associated diseases. A 30-day nutritional trial in broiler chickens (Ross 308) was conducted to investigate how specific forms of vitamin E (α- and γ-tocopherol) and their combination impact liver gene expression when oxidative susceptibility of the organism is induced by high n-3 polyunsaturated fatty acids (PUFA) intake (linseed oil). Thirty-six one-day-old male broilers were fed a diet enriched with 5% linseed oil. A control group (Cont; N = 10) was used as a reference group, Tα (N = 10) was supplemented with 67 mg/kg RRR-α-tocopherol, Tγ (N = 8) with 67 mg/kg RRR-γ-tocopherol, and Tαγ (N = 8) with a combination of 33.5 mg/kg of each tocopherol. Beside oxidative stress indicators, whole chicken genome microarray analysis was performed on liver RNA and selected differentially expressed genes were confirmed by real time quantitative PCR. α-Tocopherol alone and in combination with γ-tocopherol was able to prevent lipid oxidation, which was also supported by transcriptome analysis. The effect of γ-tocopherol was evident in the expression of genes involved in inflammatory processes and immune response, while α-tocopherol affected genes involved in lipid and cholesterol metabolism. Both isomers of vitamin E influenced the transcription of genes, which are related to improved fat oxidation and enhanced glucose sparing.
Collapse
Affiliation(s)
- Tamara Korošec
- University of Ljubljana, Biotechnical Faculty, Department of Animal Science, Groblje 3, 1230 Domžale, Slovenia
| | - Urška Tomažin
- Agricultural Institute of Slovenia, Hacquetova ulica 17, 1000 Ljubljana, Slovenia
| | - Simon Horvat
- University of Ljubljana, Biotechnical Faculty, Department of Animal Science, Groblje 3, 1230 Domžale, Slovenia.,National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia
| | - Rok Keber
- Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Janez Salobir
- University of Ljubljana, Biotechnical Faculty, Department of Animal Science, Groblje 3, 1230 Domžale, Slovenia
| |
Collapse
|
34
|
Uysal E, Dokur M, Altınay S, Saygılı Eİ, Batcıoglu K, Ceylan MS, Kazımoglu H, Uyumlu BA, Karadag M. Investigation of the Effect of Milrinone on Renal Damage in an Experimental Non-Heart Beating Donor Model. J INVEST SURG 2017; 31:402-411. [DOI: 10.1080/08941939.2017.1343880] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Erdal Uysal
- Department of General Surgery, Sanko University School of Medicine, Gaziantep, Turkey
| | - Mehmet Dokur
- Emergency, Dr. Necip Fazil City Hospital, Kahramanmaras, Turkey
| | - Serdar Altınay
- Department of Pathology, Bakırköy Dr Sadi Konuk Health Application and Research Center, Istanbul, Turkey
| | - Eyup İlker Saygılı
- Department of Biochemistry, Sanko University School of Medicine, Gaziantep, Turkey
| | - Kadir Batcıoglu
- Department of Biochemistry, Inonu University Faculty of Pharmacy, Malatya, Turkey
| | | | - Hatem Kazımoglu
- Department of Urology, Sanko Universitesi Tip Fakultesi, Gaziantep, Turkey
| | | | - Mehmet Karadag
- Biostatistic and Medical Informatics, Health Sciences Institute, Inonu University, Malatya, Turkey
| |
Collapse
|
35
|
Jensen SB, Käkelä M, Jødal L, Moisio O, Alstrup AKO, Jalkanen S, Roivainen A. Exploring the radiosynthesis and in vitro characteristics of [ 68 Ga]Ga-DOTA-Siglec-9. J Labelled Comp Radiopharm 2017; 60:439-449. [PMID: 28556976 DOI: 10.1002/jlcr.3525] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/04/2017] [Accepted: 05/23/2017] [Indexed: 12/12/2022]
Abstract
Vascular adhesion protein 1 is a leukocyte homing-associated glycoprotein, which upon inflammation rapidly translocates from intracellular sources to the endothelial cell surface. It has been discovered that the cyclic peptide residues 283-297 of sialic acid-binding IgG-like lectin 9 (Siglec-9) "CARLSLSWRGLTLCPSK" bind to vascular adhesion protein 1 and hence makes the radioactive analogues of this compound ([68 Ga]Ga-DOTA-Siglec-9) interesting as a noninvasive visualizing marker of inflammation. Three different approaches to the radiosynthesis of [68 Ga]Ga-DOTA-Siglec-9 are presented and compared with previously published methods. A simple, robust radiosynthesis of [68 Ga]Ga-DOTA-Siglec-9 with a yield of 62% (non decay-corrected) was identified, and it had a radiochemical purity >98% and a specific radioactivity of 35 MBq/nmol. Furthermore, the protein binding and stability of [68 Ga]Ga-DOTA-Siglec-9 were analyzed in vitro in mouse, rat, rabbit, pig, and human plasma and compared with in vivo pig results. The plasma in vitro protein binding of [68 Ga]Ga-DOTA-Siglec-9 was the lowest in the pig followed by rabbit, human, rat, and mouse. It was considerably higher in the in vivo pig experiments. The in vivo stability in pigs was lower than the in vitro stability. Despite considerable species differences, the observed characteristics of [68 Ga]Ga-DOTA-Siglec-9 are suitable as a positron emission tomography tracer.
Collapse
Affiliation(s)
- Svend B Jensen
- Department of Nuclear Medicine, Aalborg University Hospital, Denmark.,Department of Chemistry and Biosciences, Aalborg University, Aalborg, Denmark
| | - Meeri Käkelä
- Turku PET Centre, University of Turku, Turku, Finland
| | - Lars Jødal
- Department of Nuclear Medicine, Aalborg University Hospital, Denmark.,Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark.,Department of Veterinary Disease Biology, University of Copenhagen, Copenhagen, Denmark
| | - Olli Moisio
- Turku PET Centre, University of Turku, Turku, Finland
| | - Aage K O Alstrup
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Sirpa Jalkanen
- MediCity Research Laboratory and Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland.,Turku Centre for Disease Modelling, University of Turku, Turku, Finland
| |
Collapse
|
36
|
Zhang Y, Yi W, Yao J, Yu X, Qian C, Hu Z. Hypoxia serves a key function in the upregulated expression of vascular adhesion protein‑1 in vitro and in a rat model of hemorrhagic shock. Mol Med Rep 2017. [PMID: 28627649 PMCID: PMC5562078 DOI: 10.3892/mmr.2017.6727] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hemorrhagic shock following major trauma results in mortality, but the function of vascular adhesion protein-1 (VAP-1), implicated in intracranial hemorrhage, remains unknown. This study aimed to determine whether expression of the AOC3 gene and its encoded protein, VAP-1, is altered by hypoxia. Rat hepatic sinusoidal endothelial cells (RHSECs) and rat intestinal microvascular endothelial cells (RIMECs) were transduced with a viral vector carrying AOC3, and AOC3 mRNA expression levels were measured by reverse transcription-quantitative polymerase chain reaction. VAP-1 protein expression levels were measured by western blot analysis and compared between normoxic and hypoxic conditions. Following this, AOC3 mRNA and VAP-1 protein expression levels in hepatic and intestinal tissues were assessed in a rat model of hemorrhagic shock with or without fluid resuscitation; and serum semicarbazide-sensitive amine oxidase (SSAO) activity was measured by fluorometric assays. The effects of 2-bromoethylamine (2-BEA) on AOC3/VAP-1 levels and 24 h survival were investigated. AOC3 mRNA and VAP-1 protein levels were increased in RHSECs and RIMECs by hypoxia, and in hepatic and intestinal tissues from rats following hemorrhagic shock. Hypoxia increased serum SSAO activity in these animals. 2-BEA reduced AOC3 mRNA and VAP-1 protein levels in hepatic and intestinal tissues from rats following hemorrhagic shock, and appeared to improve survival in animals not receiving resuscitation following hemorrhagic shock. In conclusion, hemorrhagic shock upregulates AOC3/VAP-1 expressions, and this potentially occurs via hypoxia. Therefore, inhibition of VAP-1 may be beneficial in the setting of hemorrhagic shock. Further studies are required to confirm these findings and to establish whether VAP-1 may be a valid target for the development of novel therapies for hemorrhagic shock.
Collapse
Affiliation(s)
- Yuxing Zhang
- Department of General Surgery, Navy General Hospital, Beijing 100048, P.R. China
| | - Wei Yi
- Department of General Surgery, China People's Liberation Army No. 94 Hospital, Nanchang, Jiangxi 330002, P.R. China
| | - Jun Yao
- Department of General Surgery, Shanghai Changzheng Hospital, Shanghai 200003, P.R. China
| | - Xiaojun Yu
- Department of Gastroenterological Surgery, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Cheng Qian
- Department of General Surgery, Huzhou Maternity & Child Care Hospital, Huzhou, Zhejiang 313000, P.R. China
| | - Zhiqian Hu
- Department of General Surgery, Shanghai Changzheng Hospital, Shanghai 200003, P.R. China
| |
Collapse
|
37
|
Seo K, Choi JW, Kim DW, Han D, Noh SJ, Jung HS. Aminophylline Effect on Renal Ischemia-Reperfusion Injury in Mice. Transplant Proc 2017; 49:358-365. [PMID: 28219599 DOI: 10.1016/j.transproceed.2016.11.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/01/2016] [Accepted: 11/16/2016] [Indexed: 01/25/2023]
Abstract
BACKGROUND Aminophylline increases the intracellular concentration of cAMP and exerts an anti-inflammatory effect. The aim of this study was to investigate the effect of aminophylline on renal ischemia-reperfusion (I/R) injury in mice. METHODS Thirty C57BL/6 mice were divided into 3 groups. In the sham group (group S, n = 10), only right nephrectomy was performed. In the control group (group C, n = 10), after right nephrectomy, the mice were subjected to 30 minutes of left renal ischemia. In the aminophylline group (group A, n = 10), an intraperitoneal injection of aminophylline (5 mg/kg) was performed before renal ischemia. Twenty-four hours after reperfusion, the mice were euthanized, and plasma and kidney samples were obtained to analyze the serum creatinine, renal histology, and expression levels of nuclear factor-kappa B (NF-kB) and pro-inflammatory cytokines. RESULTS The serum creatinine concentration in group C was markedly elevated at 24 hours after reperfusion. Aminophylline treatment significantly reduced serum creatinine, compared with group C. Aminophylline also reduced the histological evidence of renal damage. The expression levels of NF-kB, tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1 (MCP-1), macrophage inflammatory protein-2 (MIP-2), and intercellular adhesion molecule-1 (ICAM-1) mRNA were significantly increased in group C (P < .001). Group A showed lower expression of NF-kB, TNF-α, MCP-1, MIP-2, and ICAM-1 mRNA than group C (P < .01). CONCLUSIONS Aminophylline treatment improved the renal function and indexes of renal inflammation, which suggests that it provided reno-protection against renal I/R injury.
Collapse
Affiliation(s)
- K Seo
- Department of Anesthesiology and Pain Medicine, St Vincent's Hospital, The Catholic University of Korea, Seoul, Korea
| | - J W Choi
- Department of Anesthesiology and Pain Medicine, St Vincent's Hospital, The Catholic University of Korea, Seoul, Korea
| | - D-W Kim
- Department of Anesthesiology and Pain Medicine, St Vincent's Hospital, The Catholic University of Korea, Seoul, Korea
| | - D Han
- Department of Anesthesiology and Pain Medicine, St Vincent's Hospital, The Catholic University of Korea, Seoul, Korea
| | - S J Noh
- The Research Institute of Medical Science, St Vincent's Hospital, The Catholic University of Korea, Seoul, Korea
| | - H S Jung
- Department of Anesthesiology and Pain Medicine, St Vincent's Hospital, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
38
|
Leukocyte trafficking-associated vascular adhesion protein 1 is expressed and functionally active in atherosclerotic plaques. Sci Rep 2016; 6:35089. [PMID: 27731409 PMCID: PMC5059718 DOI: 10.1038/srep35089] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/23/2016] [Indexed: 12/15/2022] Open
Abstract
Given the important role of inflammation and the potential association of the leukocyte trafficking-associated adhesion molecule vascular adhesion protein 1 (VAP-1) with atherosclerosis, this study examined whether functional VAP-1 is expressed in atherosclerotic lesions and, if so, whether it could be targeted by positron emission tomography (PET). First, immunohistochemistry revealed that VAP-1 localized to endothelial cells of intra-plaque neovessels in human carotid endarterectomy samples from patients with recent ischemic symptoms. In low-density lipoprotein receptor-deficient mice expressing only apolipoprotein B100 (LDLR-/-ApoB100/100), VAP-1 was expressed on endothelial cells lining inflamed atherosclerotic lesions; normal vessel walls in aortas of C57BL/6N control mice were VAP-1-negative. Second, we discovered that the focal uptake of VAP-1 targeting sialic acid-binding immunoglobulin-like lectin 9 based PET tracer [68Ga]DOTA-Siglec-9 in atherosclerotic plaques was associated with the density of activated macrophages (r = 0.58, P = 0.022). As a final point, we found that the inhibition of VAP-1 activity with small molecule LJP1586 decreased the density of macrophages in inflamed atherosclerotic plaques in mice. Our results suggest for the first time VAP-1 as a potential imaging target for inflamed atherosclerotic plaques, and corroborate VAP-1 inhibition as a therapeutic approach in the treatment of atherosclerosis.
Collapse
|
39
|
Trivedi PJ, Bruns T, Ward S, Mai M, Schmidt C, Hirschfield GM, Weston CJ, Adams DH. Intestinal CCL25 expression is increased in colitis and correlates with inflammatory activity. J Autoimmun 2016; 68:98-104. [PMID: 26873648 PMCID: PMC4803021 DOI: 10.1016/j.jaut.2016.01.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/29/2016] [Accepted: 01/31/2016] [Indexed: 12/13/2022]
Abstract
CCL25-mediated activation of CCR9 is critical for mucosal lymphocyte recruitment to the intestine. In immune-mediated liver injury complicating inflammatory bowel disease, intrahepatic activation of this pathway allows mucosal lymphocytes to be recruited to the liver, driving hepatobiliary destruction in primary sclerosing cholangitis (PSC). However, in mice and healthy humans CCL25 expression is restricted to the small bowel, whereas few data exist on activation of this pathway in the inflamed colon despite the vast majority of PSC patients having ulcerative colitis. Herein, we show that colonic CCL25 expression is not only upregulated in patients with active colitis, but strongly correlates with endoscopic Mayo score and mucosal TNFα expression. Moreover, approximately 90% (CD4(+)) and 30% (CD8(+)) of tissue-infiltrating T-cells in colitis were identified as CCR9(+) effector lymphocytes, compared to <10% of T-cells being CCR9(+) in normal colon. Sorted CCR9(+) lymphocytes also demonstrated enhanced cellular adhesion to stimulated hepatic sinusoidal endothelium compared with their CCR9(-) counterparts when under flow. Collectively, these results suggest that CCR9/CCL25 interactions are not only involved in colitis pathogenesis but also correlate with colonic inflammatory burden; further supporting the existence of overlapping mucosal lymphocyte recruitment pathways between the inflamed colon and liver.
Collapse
Affiliation(s)
- Palak J Trivedi
- National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit (BRU), Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom; Liver Unit, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Tony Bruns
- National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit (BRU), Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom; Dept. of Internal Medicine IV, University Hospital Jena, Jena, Germany; Center for Sepsis Control and Care, University Hospital Jena, Jena, Germany
| | - Stephen Ward
- National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit (BRU), Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom; Department of General and Colorectal Surgery, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Martina Mai
- Dept. of Internal Medicine IV, University Hospital Jena, Jena, Germany; Center for Sepsis Control and Care, University Hospital Jena, Jena, Germany
| | - Carsten Schmidt
- Dept. of Internal Medicine IV, University Hospital Jena, Jena, Germany
| | - Gideon M Hirschfield
- National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit (BRU), Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom; Liver Unit, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Chris J Weston
- National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit (BRU), Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - David H Adams
- National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit (BRU), Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom; Liver Unit, Queen Elizabeth Hospital, Birmingham, United Kingdom.
| |
Collapse
|
40
|
Knolle PA. Staying local-antigen presentation in the liver. Curr Opin Immunol 2016; 40:36-42. [PMID: 26974478 DOI: 10.1016/j.coi.2016.02.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 01/26/2016] [Accepted: 02/24/2016] [Indexed: 12/15/2022]
Abstract
The liver is known as organ with unique immune competence. Besides its unique microenvironment that is determined by gut-derived portal venous blood constituents and the presence of enzymes with immune regulatory properties, liver antigen presenting cell populations regulate antigen-specific immunity in a local fashion. In addition to bone marrow-derived dendritic cells and myeloid cells such as macrophages and monocytes, also truly liver-resident cell populations function as antigen presenting cells such as liver sinusoidal endothelial cells and hepatocytes. The functional outcome of antigen-presentation by these cell populations is diverse and ranges from generation of regulatory CD4 cells, to induction of memory CD8 T cells or deletional tolerance, which generates a complex network of antigen-presenting cells that determines hepatic immune regulation and local immune surveillance against viral infection.
Collapse
Affiliation(s)
- Percy A Knolle
- Institute of Molecular Immunology and Experimental Oncology Technische Universität München, Germany.
| |
Collapse
|
41
|
Abstract
When cholestasis occurs in patients receiving total parenteral nutrition, it is the result of many pathogenic pathways converging on the hepatic acinus. The result may be a temporary rise in liver function tests. The resulting fibrosis, portal hypertension, and jaundice are hallmarks of type 3 intestinal-associated liver disease to which children are more susceptible than adults. The key to prevention is in identifying high-risk scenarios, meticulous monitoring, and personalized prescription of parenteral nutrition solutions combined with an active approach in reducing the impact of inflammatory events when they occur by prompt use of antibiotics and line locks.
Collapse
Affiliation(s)
- Sue V Beath
- The Liver Unit, Birmingham Children's Hospital, Steelhouse Lane, Birmingham, West Midlands, B4 6NH, UK.
| | - Deirdre A Kelly
- The Liver Unit, Birmingham Children's Hospital, Steelhouse Lane, Birmingham, West Midlands, B4 6NH, UK
| |
Collapse
|
42
|
Pannecoeck R, Serruys D, Benmeridja L, Delanghe JR, van Geel N, Speeckaert R, Speeckaert MM. Vascular adhesion protein-1: Role in human pathology and application as a biomarker. Crit Rev Clin Lab Sci 2015; 52:284-300. [PMID: 26287391 DOI: 10.3109/10408363.2015.1050714] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Vascular adhesion protein-1 (VAP-1) is a member of the copper-containing amine oxidase/semicarbazide-sensitive amine oxidase (AOC/SSAO) enzyme family. SSAO enzymes catalyze oxidative deamination of primary amines, which results in the production of the corresponding aldehyde, hydrogen peroxide and ammonium. VAP-1 is continuously expressed as a transmembrane glycoprotein in the vascular wall during development and facilitates the accumulation of inflammatory cells into the inflamed environment in concert with other leukocyte adhesion molecules. The soluble form of VAP-1 is released into the circulation mainly from vascular endothelial cells. Over- and under-expression of sVAP-1 result in alterations of the reported reaction product levels, which are involved in the pathogenesis of multiple human diseases. The combination of enzymatic and adhesion capacities as well as its strong association with inflammatory pathologies makes VAP-1 an interesting therapeutic target for drug discovery. In this article, we will review the general characteristics and biological functions of VAP-1, focusing on its important role as a prognostic biomarker in human pathologies. In addition, the potential therapeutic application of VAP-1 inhibitors will be discussed.
Collapse
Affiliation(s)
| | | | | | | | - Nanja van Geel
- c Department of Dermatology , Ghent University Hospital , Gent , Belgium
| | | | | |
Collapse
|
43
|
Weston CJ, Shepherd EL, Claridge LC, Rantakari P, Curbishley SM, Tomlinson JW, Hubscher SG, Reynolds GM, Aalto K, Anstee QM, Jalkanen S, Salmi M, Smith DJ, Day CP, Adams DH. Vascular adhesion protein-1 promotes liver inflammation and drives hepatic fibrosis. J Clin Invest 2014; 125:501-20. [PMID: 25562318 DOI: 10.1172/jci73722] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 11/13/2014] [Indexed: 12/15/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) encompasses a range of manifestations, including steatosis and cirrhosis. Progressive disease is characterized by hepatic leukocyte accumulation in the form of steatohepatitis. The adhesion molecule vascular adhesion protein-1 (VAP-1) is a membrane-bound amine oxidase that promotes leukocyte recruitment to the liver, and the soluble form (sVAP-1) accounts for most circulating monoamine oxidase activity, has insulin-like effects, and can initiate oxidative stress. Here, we determined that hepatic VAP-1 expression is increased in patients with chronic liver disease and that serum sVAP-1 levels are elevated in patients with NAFLD compared with those in control individuals. In 4 murine hepatic injury models, an absence or blockade of functional VAP-1 reduced inflammatory cell recruitment to the liver and attenuated fibrosis. Moreover, disease was reduced in animals expressing a catalytically inactive form of VAP-1, implicating enzyme activity in the disease pathogenesis. Within the liver, hepatic stromal cells expressed functional VAP-1, and evaluation of cultured cells revealed that sVAP-1 promotes leukocyte migration through catalytic generation of ROS, which depended on VAP-1 enzyme activity. VAP-1 enhanced stromal cell spreading and wound closure and modulated expression of profibrotic genes. Together, these results link the amine oxidase activity of VAP-1 with hepatic inflammation and fibrosis and suggest that targeting VAP-1 has therapeutic potential for NAFLD and other chronic fibrotic liver diseases.
Collapse
|
44
|
Karim S, Liaskou E, Fear J, Garg A, Reynolds G, Claridge L, Adams DH, Newsome PN, Lalor PF. Dysregulated hepatic expression of glucose transporters in chronic disease: contribution of semicarbazide-sensitive amine oxidase to hepatic glucose uptake. Am J Physiol Gastrointest Liver Physiol 2014; 307:G1180-90. [PMID: 25342050 PMCID: PMC4269679 DOI: 10.1152/ajpgi.00377.2013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Insulin resistance is common in patients with chronic liver disease (CLD). Serum levels of soluble vascular adhesion protein-1 (VAP-1) are also increased in these patients. The amine oxidase activity of VAP-1 stimulates glucose uptake via translocation of transporters to the cell membrane in adipocytes and smooth muscle cells. We aimed to document human hepatocellular expression of glucose transporters (GLUTs) and to determine if VAP-1 activity influences receptor expression and hepatic glucose uptake. Quantitative PCR and immunocytochemistry were used to study human liver tissue and cultured cells. We also used tissue slices from humans and VAP-1-deficient mice to assay glucose uptake and measure hepatocellular responses to stimulation. We report upregulation of GLUT1, -3, -5, -6, -7, -8, -9, -10, -11, -12, and -13 in CLD. VAP-1 expression and enzyme activity increased in disease, and provision of substrate to hepatic VAP-1 drives hepatic glucose uptake. This effect was sensitive to inhibition of VAP-1 and could be recapitulated by H2O2. VAP-1 activity also altered expression and subcellular localization of GLUT2, -4, -9, -10, and -13. Therefore, we show, for the first time, alterations in hepatocellular expression of glucose and fructose transporters in CLD and provide evidence that the semicarbazide-sensitive amine oxidase activity of VAP-1 modifies hepatic glucose homeostasis and may contribute to patterns of GLUT expression in chronic disease.
Collapse
Affiliation(s)
- Sumera Karim
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and
| | - Evaggelia Liaskou
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and
| | - Janine Fear
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and
| | - Abhilok Garg
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and
| | - Gary Reynolds
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and
| | - Lee Claridge
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and
| | - David H. Adams
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and ,2Liver and Hepatobiliary Unit, Queen Elizabeth Hospital, Edgbaston, Birmingham, United Kingdom
| | - Philip N. Newsome
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and ,2Liver and Hepatobiliary Unit, Queen Elizabeth Hospital, Edgbaston, Birmingham, United Kingdom
| | - Patricia F. Lalor
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and
| |
Collapse
|
45
|
Aalto K, Havulinna AS, Jalkanen S, Salomaa V, Salmi M. Soluble vascular adhesion protein-1 predicts incident major adverse cardiovascular events and improves reclassification in a finnish prospective cohort study. ACTA ACUST UNITED AC 2014; 7:529-35. [PMID: 24850810 DOI: 10.1161/circgenetics.113.000543] [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: 12/30/2022]
Abstract
BACKGROUND Vascular adhesion protein-1 (VAP-1) associates to subclinical atherosclerotic manifestations in young people, but its association to incident major adverse cardiovascular events (MACEs) and cardiovascular mortality in a general population is not known. METHODS AND RESULTS We used a newly developed ELISA to measure soluble VAP-1 (sVAP-1) levels in 2775 participants (mean age, 60 years) from a prospective cohort study (the FINRISK 2002). During a mean follow-up of 9 years, 265 participants underwent a MACE, and these participants had higher levels of sVAP-1 than those without MACE (868 ng/mL and 824 ng/mL, respectively, P<0.001). In multivariate-adjusted Cox proportional hazard model including traditional Framingham risk factors (age, sex, systolic blood pressure, cholesterol, high-density lipoprotein cholesterol, smoking, prevalent diabetes mellitus, and antihypertensive treatment), sVAP-1 independently predicted incident MACE (P=0.0046) and MACE mortality (P=0.026). The impact of sVAP-1 in predicting the 9-year absolute risk of MACE was analyzed using integrated discrimination improvement and net reclassification improvement with 10-fold cross-validation. Inclusion of sVAP-1 in the Framingham model improved integrated discrimination improvement (P=0.042), and the clinical net reclassification improvement by correctly reclassifying 9% (P=0.0019) of people in the intermediate risk (5%-20%) group. CONCLUSIONS sVAP-1 associated with increased risk of MACE and MACE mortality in people aged >50 years without prior MACE, and inclusion of sVAP-1 in the risk prediction model improved the clinical net reclassification improvement of incident MACE. Thus, sVAP-1 may be a potential new biomarker for cardiovascular diseases.
Collapse
Affiliation(s)
- Kristiina Aalto
- From the MediCity Research Laboratory, and Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland (K.A., S.J., M.S.), and Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku and Helsinki, Finland (A.S.H., S.J., V.S., M.S.)
| | - Aki S Havulinna
- From the MediCity Research Laboratory, and Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland (K.A., S.J., M.S.), and Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku and Helsinki, Finland (A.S.H., S.J., V.S., M.S.)
| | - Sirpa Jalkanen
- From the MediCity Research Laboratory, and Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland (K.A., S.J., M.S.), and Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku and Helsinki, Finland (A.S.H., S.J., V.S., M.S.)
| | - Veikko Salomaa
- From the MediCity Research Laboratory, and Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland (K.A., S.J., M.S.), and Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku and Helsinki, Finland (A.S.H., S.J., V.S., M.S.)
| | - Marko Salmi
- From the MediCity Research Laboratory, and Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland (K.A., S.J., M.S.), and Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku and Helsinki, Finland (A.S.H., S.J., V.S., M.S.).
| |
Collapse
|
46
|
Ectoenzymes in leukocyte migration and their therapeutic potential. Semin Immunopathol 2014; 36:163-76. [PMID: 24638888 DOI: 10.1007/s00281-014-0417-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/19/2014] [Indexed: 02/07/2023]
Abstract
Inflammation causes or accompanies a huge variety of diseases. Migration of leukocytes from the blood into the tissues, in the tissues, and from the tissues to lymphatic vasculature is crucial in the formation and resolution of inflammatory infiltrates. In addition to classical adhesion and activation molecules, several other molecules are known to contribute to the leukocyte traffic. Several of them belong to ectoenzymes, which are cell surface molecules having catalytically active sites outside the cell. We will review here how several ectoenzymes present on leukocytes or endothelial cell surface function as adhesins and/or modulate the extravasation cascade through their enzymatic activities. Moreover, their therapeutic potential as immune modulators in different experimental inflammation models and in clinical trials will be discussed.
Collapse
|
47
|
Jung HS, Joo JD, Kim DW, In JH, Roh M, Jeong JT, Noh SJ, Choi JW. Effect of milrinone on the inflammatory response and NF-kB activation in renal ischemia-reperfusion injury in mice. Korean J Anesthesiol 2014; 66:136-42. [PMID: 24624272 PMCID: PMC3948441 DOI: 10.4097/kjae.2014.66.2.136] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 10/15/2013] [Accepted: 10/16/2013] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Milrinone increases intracellular adenosine 3',5'-cyclic monophosphate concentration and enhances vascular relaxation. Nuclear factor-kappa B (NF-kB) plays a key role in inflammatory responses during ischemia-reperfusion (I/R) injury. We aimed to investigate the effect of milrinone on the inflammatory responses and NF-kB activation in renal I/R injury in mice. METHODS Thirty C57BL/6 mice were allocated into 3 groups. In group S (n = 10), only right nephrectomy was done. In group C (n = 10), the left kidney was subjected to 30 min of ischemia after right nephrectomy. In group M (n = 10), milrinone (5 µg/kg) was administered before ischemia. After 24 hours of reperfusion, the serum creatinine was measured, kidney samples were obtained for histology, and expressions of NF-kB and proinflammatory cytokines were analyzed. RESULTS In group C, the serum creatinine concentration was markedly elevated, compared with group S. Creatinine concentration in group M was also elevated, but it was significantly lower than that in group C. Histologic evidence of renal damage was severe in group C, but it was improved in group M. In groups C and M, expression of NF-kB, tumor necrosis factor-α (TNF-α), intercellular adhesion molecule-1 (ICAM-1), monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-2 (MIP-2) mRNA increased significantly compared with group S (P < 0.05). But group M showed a lower expression of NF-kB, TNF-α, ICAM-1, MCP-1 and MIP-2 mRNA than group C (P < 0.05). CONCLUSIONS Milrinone treatment attenuates the renal inflammatory response and activation of NF-kB, resulting in improvement of renal function and tissue injury.
Collapse
Affiliation(s)
- Hong Soo Jung
- Department of Anesthesiology and Pain Medicine, College of Medicine, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Jin-Deok Joo
- Department of Anesthesiology and Pain Medicine, College of Medicine, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Dae-Woo Kim
- Department of Anesthesiology and Pain Medicine, College of Medicine, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Jang Hyeok In
- Department of Anesthesiology and Pain Medicine, College of Medicine, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Misun Roh
- Department of Anesthesiology and Pain Medicine, College of Medicine, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Jong-Tae Jeong
- Department of Anesthesiology and Pain Medicine, College of Medicine, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Seung June Noh
- The Research Institute of Medical Science, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Jin Woo Choi
- Department of Anesthesiology and Pain Medicine, College of Medicine, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| |
Collapse
|
48
|
Lin HL, Yen HW, Hsieh SL, An LM, Shen KP. Low-dose aspirin ameliorated hyperlipidemia, adhesion molecule, and chemokine production induced by high-fat diet in Sprague-Dawley rats. Drug Dev Res 2013; 75:97-106. [PMID: 24648214 DOI: 10.1002/ddr.21159] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 11/06/2013] [Indexed: 11/07/2022]
Abstract
In this study the effects of low-dose aspirin (5 mg/kg) on adhesion molecule and chemokine expression in a hyperlipidemic rat model. Six-week-old Sprague-Dawley (SD) rats were assigned to two control groups receiving either a regular diet or high-fat diet (HFD) and a treatment group fed HFD with 5 mg/kg aspirin for a 10-week period. Compared with the regular diet control group, the HFD control group had higher body weight, lower levels of high-density lipoprotein, higher concentrations of insulin, triglyceride, total cholesterol, and low-density lipoprotein, but no differences in blood glucose and glycated hemoglobin. The prothrombin time (PT) and activated partial thromboplastin time (aPTT) were clearly shortened in the HFD group. That group also had increased expression of intercellular adhesion molecule-1 (ICAM-1), ICAM-2, ICAM-3, vascular cell adhesion molecule (VCAM), platelet endothelial cell adhesion molecule (PECAM) and P-selectin in platelets and vascular adhesion protein-1 in lymphocyte and in aorta increased expressions of ICAM-1, ICAM-2, ICAM-3, VCAM, PECAM, E-selectin, monocyte chemoattractant protein-1 (MCP-1) and CCR2. The HFD rats also had increased PKCα, IκB kinase α (IKKα), p65, mitogen-activated protein kinases (MAPKs) (p38, c-Jun N-terminal kinases 1, extracellular signal-regulated kinase 1/2), and their phosphorylated forms. Low-dose aspirin improved HFD-induced hyperinsulinemia and hyperlipidemia, recovered PT and aPTT, inhibited upregulation of adhesion molecules and chemokines and reduced expression of PKCα, IKKα, p65, and MAPKs. Low-dose aspirin ameliorates HFD-induced hyperlipidemia and hyperinsulinemia, and prevents HFD-induced expression of adhesion molecules and chemokine formation.
Collapse
Affiliation(s)
- Hui-Li Lin
- Department of Food Science and Nutrition, Meiho University, Pingtung, Taiwan
| | | | | | | | | |
Collapse
|
49
|
Foot JS, Yow TT, Schilter H, Buson A, Deodhar M, Findlay AD, Guo L, McDonald IA, Turner CI, Zhou W, Jarolimek W. PXS-4681A, a potent and selective mechanism-based inhibitor of SSAO/VAP-1 with anti-inflammatory effects in vivo. J Pharmacol Exp Ther 2013; 347:365-74. [PMID: 23943052 DOI: 10.1124/jpet.113.207613] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Semicarbazide-sensitive amine oxidase (SSAO), also known as vascular adhesion protein-1 (VAP-1), is a member of the copper-dependent amine oxidase family that is associated with various forms of inflammation and fibrosis. To investigate the therapeutic potential of SSAO/VAP-1 inhibition, potent and selective inhibitors with drug-like properties are required. PXS-4681A [(Z)-4-(2-(aminomethyl)-3-fluoroallyloxy)benzenesulfonamide hydrochloride] is a mechanism-based inhibitor of enzyme function with a pharmacokinetic and pharmacodynamic profile that ensures complete, long-lasting inhibition of the enzyme after a single low dose in vivo. PXS-4681A irreversibly inhibits the enzyme with an apparent Ki of 37 nM and a kinact of 0.26 min(-1) with no observed turnover in vitro. It is highly selective for SSAO/VAP-1 when profiled against related amine oxidases, ion channels, and seven-transmembrane domain receptors, and is superior to previously reported inhibitors. In mouse models of lung inflammation and localized inflammation, dosing of this molecule at 2 mg/kg attenuates neutrophil migration, tumor necrosis factor-α, and interleukin-6 levels. These results demonstrate the drug-like properties of PXS-4681A and its potential use in the treatment of inflammation.
Collapse
|
50
|
Wong MYW, Saad S, Pollock C, Wong MG. Semicarbazide-sensitive amine oxidase and kidney disease. Am J Physiol Renal Physiol 2013; 305:F1637-44. [PMID: 24173357 DOI: 10.1152/ajprenal.00416.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
With better understanding of the molecular mechanisms underpinning chronic kidney disease, the roles of inflammation and fibrosis are becoming increasingly inseparable. The progression of renal disease is characterized by pathomorphological changes that consist of early inflammatory responses followed by tubulointerstitial fibrosis, tubular atrophy, and glomerular and vascular sclerosis. Currently available therapies that reduce hypertension, proteinuria, hyperglycemia, and interruption of the renin-angiotensin-aldosterone system are at best only partially effective. Hence, there remains a need to explore agents targeting nonrenin-angiotensin-aldosterone system pathways. In this review, we discuss mechanistic aspects in the physiological and pathological role of semicarbazide-sensitive amine oxidase, a protein enzyme involved in cellular trafficking and inflammation, with respect to the kidney. We explore the evidence for the use of semicarbazide-sensitive amine oxidase inhibitors as potential agents in renal fibrosis to delay the onset and progression of chronic kidney disease.
Collapse
Affiliation(s)
- May Y W Wong
- Kolling Institute of Medical Research, Level 7 Kolling Bldg., Royal North Shore Hospital, St Leonards 2065, NSW, Australia.
| | | | | | | |
Collapse
|