1
|
Gao F, Mu W, Fan J, Shen J. β-arrestin2 promotes angiogenesis of liver sinusoidal endothelial cells through the VEGF/VEGFR2 pathway to aggravate cirrhosis. Toxicol Lett 2024; 401:1-12. [PMID: 39197505 DOI: 10.1016/j.toxlet.2024.08.011] [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: 05/11/2024] [Revised: 07/24/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024]
Abstract
Excessive extracellular matrix deposition and increased intrahepatic angiogenesis are prominent features of cirrhosis. β-arrestin2 is thought to be involved in the pathological processes of various fibrotic diseases. This study aimed to investigate the role and possible mechanism of β-arrestin2 in the angiogenesis of cirrhosis. Firstly, β-arrestin2 expression in liver tissues of cirrhotic patients was detected, and the correlation between β-arrestin2 and α-SMA, CD-31, PDGF, and VEGF indexes was analyzed. Then, after liver cirrhosis induced by CCL4 in Arrb2-KO mice (β-arrestin2 coding gene), liver histopathological changes were observed, and the expressions of α-SMA, CD-31, PDGF, VEGF, and VEGFR2 were detected. Finally, VEGF-A was used to treat human liver sinusoidal endothelial cells (LSECs) to simulate pathological conditions. After transfection with si-ARRB2, the cell activity, MDA and GSH-PX activities, cell invasion, angiogenesis, and the expressions of α-SMA, CD-31, and VEGF/VEGFR2 pathway were detected. Results showed that β-arrestin2 expression in the liver increased significantly during cirrhosis and was positively correlated with angiogenesis. In vivo, Arrb2-KO significantly inhibited fibrosis and angiogenesis in cirrhotic mice, and decreased the expressions of α-SMA, CD31, PDGF, VEGF, and VEGFR2. Studies using LSECs in vitro showed that after intervention of ARRB2, the activity of LSECs and the number of invasions and tubule formations were significantly reduced. Similarly, after transfection with si-ARRB2, the expressions of α-SMA, CD31, PDGF, VEGF, and VEGFR2 in LSECs were significantly decreased. Collectively, β-arrestin2 aggravated cirrhosis by promoting the angiogenesis of LSECs. Blocking β-arrestin2 may be an important target against angiogenesis and fibrosis in cirrhosis.
Collapse
Affiliation(s)
- Feng Gao
- Department of Interventional Therapy, Shanxi Provincial People's Hospital, Taiyuan 030012, China
| | - Wei Mu
- Department of Interventional Therapy, Shanxi Provincial People's Hospital, Taiyuan 030012, China
| | - Jiangbo Fan
- Department of Interventional Therapy, Shanxi Provincial People's Hospital, Taiyuan 030012, China
| | - Jing Shen
- Department of Interventional Therapy, Shanxi Provincial People's Hospital, Taiyuan 030012, China.
| |
Collapse
|
2
|
Jiang H, Huang X, Wang J, Zhou Y, Ren C, Zhou T, Pei J. Hepatoprotective Effect of Medicine Food Homology Flower Saffron against CCl 4-Induced Liver Fibrosis in Mice via the Akt/HIF-1α/VEGF Signaling Pathway. Molecules 2023; 28:7238. [PMID: 37959658 PMCID: PMC10648070 DOI: 10.3390/molecules28217238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/15/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023] Open
Abstract
Liver fibrosis refers to a complex inflammatory response caused by multiple factors, which is a known cause of liver cirrhosis and even liver cancer. As a valuable medicine food homology herb, saffron has been widely used in the world. Saffron is commonly used in liver-related diseases and has rich therapeutic and health benefits. The therapeutic effect is satisfactory, but its mechanism is still unclear. In order to clarify these problems, we planned to determine the pharmacological effects and mechanisms of saffron extract in preventing and treating liver fibrosis through network pharmacology analysis combined with in vivo validation experiments. Through UPLC-Q-Exactive-MS analysis, a total of fifty-six nutrients and active ingredients were identified, and nine of them were screened to predict their therapeutic targets for liver fibrosis. Then, network pharmacology analysis was applied to identify 321 targets for saffron extract to alleviate liver fibrosis. Functional and pathway enrichment analysis showed that the putative targets of saffron for the treatment of hepatic fibrosis are mainly involved in the calcium signaling pathway, the HIF-1 signaling pathway, endocrine resistance, the PI3K/Akt signaling pathway, lipid and atherosclerosis, and the cAMP signaling pathway. Based on the CCl4-induced liver fibrosis mice model, we experimentally confirmed that saffron extract can alleviate the severity and pathological changes during the progression of liver fibrosis. RT-PCR and Western blotting analysis confirmed that saffron treatment can prevent the CCl4-induced upregulation of HIF-1α, VEGFA, AKT, and PI3K, suggesting that saffron may regulate AKT/HIF-1α/VEGF and alleviate liver fibrosis.
Collapse
Affiliation(s)
- Huajuan Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China; (H.J.); (X.H.); (C.R.)
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xulong Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China; (H.J.); (X.H.); (C.R.)
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jiaxin Wang
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China;
| | - Yongfeng Zhou
- The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230031, China;
| | - Chaoxiang Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China; (H.J.); (X.H.); (C.R.)
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tao Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China; (H.J.); (X.H.); (C.R.)
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jin Pei
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China; (H.J.); (X.H.); (C.R.)
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
3
|
Deng J, Qin L, Zhou Z. Network Pharmacology and Molecular Docking Reveal the Mechanism of Isodon ternifolius (D. Don) Kudo Against Liver Fibrosis. Drug Des Devel Ther 2023; 17:2335-2351. [PMID: 37576085 PMCID: PMC10416792 DOI: 10.2147/dddt.s412818] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/18/2023] [Indexed: 08/15/2023] Open
Abstract
Aim Many studies have demonstrated the hepatoprotective or anti-fibrotic effects of Isodon ternifolius, but its pharmacological basis and mechanism remain unclear. In this study, we used in vitro models to validate the predicted results and revealed the potential mechanism of action and active ingredients through network pharmacology methods and molecular docking. Methods The chemical components of Isodon ternifolius were identified by literatures. Potential targets of Isodon ternifolius were predicted by Swiss Target Prediction. The disease targets were collected through the databases of Gene Card. Common targets of Isodon ternifolius and liver fibrosis were obtained by online tool Venny 2.1. PPI protein interaction network was obtained using String database, and target protein interaction network was drawn using Cytoscape software. Signaling pathway enrichment analysis was performed on drug-disease targets with of DAVID database. Results Twenty-one potential active ingredients and 298 potential targets were predicted by Swiss Target Prediction platform. Ninety pathways related to liver fibrosis were obtained by KEGG enrichment. The TLR4, MAPK and PI3K-Akt signaling pathways are mostly associated with liver fibrosis. Molecular docking techniques were used to validate the core target proteins TNF, Akt1, MAPK1, EGFR and TLR4 binding to the ingredients of Isodon ternifolius, which showed that a multitude of ingredients of Isodon ternifolius were able to bind to the above target proteins, especially 2α-hydroxy oleanolic acid and (-)-Lambertic acid. Our experimental validation results showed that Isodon ternifolius inhibited the activation of PI3K-Akt and ERK1/2 signaling pathways. Conclusion Through a network pharmacology approach and in vitro cell assay, we predicted and validated the active compounds of Isodon ternifolius and its potential targets for LF treatment. The results suggest that the mechanism of Isodon ternifolius treating LF by inhibiting angiogenesis may be related to the ERK1/2 and PI3K/Akt signaling pathways.
Collapse
Affiliation(s)
- Jiasheng Deng
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning, Guangxi, People’s Republic of China
| | - Le Qin
- Department of Pharmacy, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, People’s Republic of China
| | - Zhipin Zhou
- Department of Pharmacy, Liuzhou People’s Hospital, Liuzhou, Guangxi, People’s Republic of China
| |
Collapse
|
4
|
Luu RJ, Hoefler BC, Gard AL, Ritenour CR, Rogers MT, Kim ES, Coppeta JR, Cain BP, Isenberg BC, Azizgolshani H, Fajardo-Ramirez OR, García-Cardeña G, Lech MP, Tomlinson L, Charest JL, Williams C. Fibroblast activation in response to TGFβ1 is modulated by co-culture with endothelial cells in a vascular organ-on-chip platform. Front Mol Biosci 2023; 10:1160851. [PMID: 37577751 PMCID: PMC10421749 DOI: 10.3389/fmolb.2023.1160851] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/06/2023] [Indexed: 08/15/2023] Open
Abstract
Background: Tissue fibrosis is a major healthcare burden that affects various organs in the body for which no effective treatments exist. An underlying, emerging theme across organs and tissue types at early stages of fibrosis is the activation of pericytes and/or fibroblasts in the perivascular space. In hepatic tissue, it is well known that liver sinusoidal endothelial cells (EC) help maintain the quiescence of stellate cells, but whether this phenomenon holds true for other endothelial and perivascular cell types is not well studied. Methods: The goal of this work was to develop an organ-on-chip microvascular model to study the effect of EC co-culture on the activation of perivascular cells perturbed by the pro-fibrotic factor TGFβ1. A high-throughput microfluidic platform, PREDICT96, that was capable of imparting physiologically relevant fluid shear stress on the cultured endothelium was utilized. Results: We first studied the activation response of several perivascular cell types and selected a cell source, human dermal fibroblasts, that exhibited medium-level activation in response to TGFβ1. We also demonstrated that the PREDICT96 high flow pump triggered changes in select shear-responsive factors in human EC. We then found that the activation response of fibroblasts was significantly blunted in co-culture with EC compared to fibroblast mono-cultures. Subsequent studies with conditioned media demonstrated that EC-secreted factors play at least a partial role in suppressing the activation response. A Luminex panel and single cell RNA-sequencing study provided additional insight into potential EC-derived factors that could influence fibroblast activation. Conclusion: Overall, our findings showed that EC can reduce myofibroblast activation of perivascular cells in response to TGFβ1. Further exploration of EC-derived factors as potential therapeutic targets in fibrosis is warranted.
Collapse
Affiliation(s)
- Rebeccah J. Luu
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - B. Christopher Hoefler
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Ashley L. Gard
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | | | - Miles T. Rogers
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Ernest S. Kim
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Jonathan R. Coppeta
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Brian P. Cain
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Brett C. Isenberg
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Hesham Azizgolshani
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Oscar R. Fajardo-Ramirez
- Laboratory for Systems Mechanobiology, Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Guillermo García-Cardeña
- Laboratory for Systems Mechanobiology, Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | | | | | - Joseph L. Charest
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Corin Williams
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| |
Collapse
|
5
|
Han C, Leonardo TR, Romana-Souza B, Shi J, Keiser S, Yuan H, Altakriti M, Ranzer MJ, Ferri-Borgogno S, Mok SC, Koh TJ, Hong SJ, Chen L, DiPietro LA. Microfibril-associated protein 5 and the regulation of skin scar formation. Sci Rep 2023; 13:8728. [PMID: 37253753 PMCID: PMC10229580 DOI: 10.1038/s41598-023-35558-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/20/2023] [Indexed: 06/01/2023] Open
Abstract
Many factors regulate scar formation, which yields a modified extracellular matrix (ECM). Among ECM components, microfibril-associated proteins have been minimally explored in the context of skin wound repair. Microfibril-associated protein 5 (MFAP5), a small 25 kD serine and threonine rich microfibril-associated protein, influences microfibril function and modulates major extracellular signaling pathways. Though known to be associated with fibrosis and angiogenesis in certain pathologies, MFAP5's role in wound healing is unknown. Using a murine model of skin wound repair, we found that MFAP5 is significantly expressed during the proliferative and remodeling phases of healing. Analysis of existing single-cell RNA-sequencing data from mouse skin wounds identified two fibroblast subpopulations as the main expressors of MFAP5 during wound healing. Furthermore, neutralization of MFAP5 in healing mouse wounds decreased collagen deposition and refined angiogenesis without altering wound closure. In vitro, recombinant MFAP5 significantly enhanced dermal fibroblast migration, collagen contractility, and expression of pro-fibrotic genes. Additionally, TGF-ß1 increased MFAP5 expression and production in dermal fibroblasts. Our findings suggest that MFAP5 regulates fibroblast function and influences scar formation in healing wounds. Our work demonstrates a previously undescribed role for MFAP5 and suggests that microfibril-associated proteins may be significant modulators of wound healing outcomes and scarring.
Collapse
Affiliation(s)
- Chen Han
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA
| | - Trevor R Leonardo
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Bruna Romana-Souza
- Department of Histology and Embryology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Junhe Shi
- NMPA Key Laboratory for Clinical Research and Evaluation of Traditional Chinese Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shalyn Keiser
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA
| | - Heidi Yuan
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA
| | - Mohamad Altakriti
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA
| | - Matthew J Ranzer
- Department of Surgery, University of Illinois Chicago, Chicago, IL, USA
| | - Sammy Ferri-Borgogno
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samuel C Mok
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy J Koh
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL, USA
| | - Seok Jong Hong
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, IL, USA
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA.
| | - Luisa A DiPietro
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
6
|
Melaibari M, Alkreathy HM, Esmat A, Rajeh NA, Shaik RA, Alghamdi AA, Ahmad A. Anti-Fibrotic Efficacy of Apigenin in a Mice Model of Carbon Tetrachloride-Induced Hepatic Fibrosis by Modulation of Oxidative Stress, Inflammation, and Fibrogenesis: A Preclinical Study. Biomedicines 2023; 11:biomedicines11051342. [PMID: 37239014 DOI: 10.3390/biomedicines11051342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Hepatic fibrosis is a major health problem all over the world, and there is no effective treatment to cure it. Hence, the current study sought to assess the anti-fibrotic efficacy of apigenin against CCl4-induced hepatic fibrosis in mice. METHODS Forty-eight mice were put into six groups. G1: Normal Control, G2: CCl4 Control, G3: Silymarin (100 mg/kg), G4 and G5: Apigenin (2 &20 mg/Kg), G6: Apigenin alone (20 mg/Kg). Groups 2, 3, 4, and 5 were given CCl4 (0.5 mL/kg. i.p.) twice/week for six weeks. The level of AST, ALT, TC, TG, and TB in serum and IL-1β, IL-6, and TNF-α in tissue homogenates were assessed. Histological studies by H&E staining and Immunostaining of liver tissues were also performed. RESULTS The CCl4-challenged group showed increased serum AST (4-fold), ALT (6-fold), and TB (5-fold). Both silymarin and apigenin treatments significantly improved these hepatic biomarkers. The CCl4-challenged group showed reduced levels of CAT (89%), GSH (53%), and increased MDA (3-fold). Both silymarin and apigenin treatments significantly altered these oxidative markers in tissue homogenates. The CCl4-treated group showed a two-fold increase in IL-1β, IL-6, and TNF-α levels. Silymarin and apigenin treatment considerably decreased the IL-1β, IL-6, and TNF-α levels. Apigenin treatment inhibited angiogenic activity, as evidenced by a decrease in VEGF (vascular endothelial growth factor) expression in liver tissues, and a decline in vascular endothelial cell antigen expression (CD34). CONCLUSIONS Finally, these data collectively imply that apigenin may have antifibrotic properties, which may be explained by its anti-inflammatory, antioxidant, and antiangiogenic activities.
Collapse
Affiliation(s)
- Maryam Melaibari
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Huda M Alkreathy
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ahmed Esmat
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Nisreen A Rajeh
- Department of Clinical Anatomy, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Rasheed A Shaik
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Anwar A Alghamdi
- Health Information Technology Department, The Applied College, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Pharmacovigilance and Medication Safety Unit, Center of Research Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Aftab Ahmad
- Health Information Technology Department, The Applied College, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Pharmacovigilance and Medication Safety Unit, Center of Research Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
7
|
Bulatova IA, Shevlyukova TP, Shchekotova AP, Paducheva SV. Analysis of cytokine gene polymorphisms in patients with chronic diffusional liver diseases in Perm krai. EXPERIMENTAL AND CLINICAL GASTROENTEROLOGY 2022:90-95. [DOI: 10.31146/1682-8658-ecg-203-7-90-95] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Irina A. Bulatova
- University E. A. Wagner Perm State Medical University of the Ministry of Health of the Russian Federation
| | | | - Alevtina P. Shchekotova
- University E. A. Wagner Perm State Medical University of the Ministry of Health of the Russian Federation
| | | |
Collapse
|
8
|
Lin Y, Dong M, Liu Z, Xu M, Huang Z, Liu H, Gao Y, Zhou W. A strategy of vascular-targeted therapy for liver fibrosis. Hepatology 2022; 76:660-675. [PMID: 34940991 PMCID: PMC9543235 DOI: 10.1002/hep.32299] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS No effective treatments are available for liver fibrosis. Angiogenesis is deeply involved in liver fibrogenesis. However, current controversial results suggest it is difficult to treat liver fibrosis through vascular targeting. There are three different microvessels in liver: portal vessels, liver sinusoids, and central vessels. The changes and roles for each of the three different vessels during liver fibrogenesis are unclear. We propose that they play different roles during liver fibrogenesis, and a single vascular endothelial cell (EC) regulator is not enough to fully regulate these three vessels to treat liver fibrosis. Therefore, a combined regulation of multiple different EC regulatory signaling pathway may provide new strategies for the liver fibrosis therapy. Herein, we present a proof-of-concept strategy by combining the regulation of leukocyte cell-derived chemotaxin 2 (LECT2)/tyrosine kinase with immunoglobulin-like and epidermal growth factor-like domains 1 signaling with that of vascular endothelial growth factor (VEGF)/recombinant VEGF (rVEGF) signaling. APPROACH AND RESULTS The CCl4 -induced mouse liver fibrosis model and NASH model were both used. During fibrogenesis, vascular changes occurred at very early stage, and different liver vessels showed different changes and played different roles: decreased portal vessels, increased sinusoid capillarization and the increased central vessels the increase of portal vessels alleviates liver fibrosis, the increase of central vessels aggravates liver fibrosis, and the increase of sinusoid capillarization aggravates liver fibrosis. The combinational treatment of adeno-associated viral vector serotype 9 (AAV9)-LECT2-short hairpin RNA (shRNA) and rVEGF showed improved therapeutic effects, but it led to serious side effects. The combination of AAV9-LECT2-shRNA and bevacizumab showed both improved therapeutic effects and decreased side effects. CONCLUSIONS Liver vascular changes occurred at very early stage of fibrogenesis. Different vessels play different roles in liver fibrosis. The combinational treatment of AAV9-LECT2-shRNA and bevacizumab could significantly improve the therapeutic effects on liver fibrosis.
Collapse
Affiliation(s)
- Yuan Lin
- Department of PathologyShunde HospitalSouthern Medical University (The First People’s Hospital of Shunde Foshan)FoshanChina,State Key Laboratory of Organ Failure ResearchDepartment of PathologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Meng‐Qi Dong
- State Key Laboratory of Organ Failure ResearchDepartment of PathologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Zhi‐Min Liu
- State Key Laboratory of Organ Failure ResearchDepartment of PathologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Meng Xu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal TumorNanfang HospitalFirst Clinical Medical CollegeSouthern Medical UniversityGuangzhouChina
| | - Zhi‐Hao Huang
- State Key Laboratory of Organ Failure ResearchDepartment of PathologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Hong‐Juan Liu
- Department of BioinformationSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Yi Gao
- General Surgery CenterDepartment of Hepatobiliary Surgery IIGuangdong ProvincialResearch Center for Artificial Organ and Tissue EngineeringGuangzhou Clinical Research and Transformation Center for Artificial LiverInstitute of Regenerative MedicineZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Wei‐Jie Zhou
- Department of PathologyShunde HospitalSouthern Medical University (The First People’s Hospital of Shunde Foshan)FoshanChina,State Key Laboratory of Organ Failure ResearchDepartment of PathologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina,Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal TumorNanfang HospitalFirst Clinical Medical CollegeSouthern Medical UniversityGuangzhouChina,General Surgery CenterDepartment of Hepatobiliary Surgery IIGuangdong ProvincialResearch Center for Artificial Organ and Tissue EngineeringGuangzhou Clinical Research and Transformation Center for Artificial LiverInstitute of Regenerative MedicineZhujiang HospitalSouthern Medical UniversityGuangzhouChina,Microbiome Medicine CenterZhujiang HospitalSouthern Medical UniversityGuangzhouChina,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| |
Collapse
|
9
|
Van Loy T, De Jonghe S, Castermans K, Dheedene W, Stoop R, Verschuren L, Versele M, Chaltin P, Luttun A, Schols D. Stimulation of the atypical chemokine receptor 3 (ACKR3) by a small-molecule agonist attenuates fibrosis in a preclinical liver but not lung injury model. Cell Mol Life Sci 2022; 79:293. [PMID: 35562519 PMCID: PMC9106635 DOI: 10.1007/s00018-022-04317-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/12/2022] [Accepted: 04/19/2022] [Indexed: 11/03/2022]
Abstract
Atypical chemokine receptor 3 (ACKR3, formerly CXC chemokine receptor 7) is a G protein-coupled receptor that recruits β-arrestins, but is devoid of functional G protein signaling after receptor stimulation. In preclinical models of liver and lung fibrosis, ACKR3 was previously shown to be upregulated after acute injury in liver sinusoidal and pulmonary capillary endothelial cells, respectively. This upregulation was linked with a pro-regenerative and anti-fibrotic role for ACKR3. A recently described ACKR3-targeting small molecule agonist protected mice from isoproterenol-induced cardiac fibrosis. Here, we aimed to evaluate its protective role in preclinical models of liver and lung fibrosis. After confirming its in vitro pharmacological activity (i.e., ACKR3-mediated β-arrestin recruitment and receptor binding), in vivo administration of this ACKR3 agonist led to increased mouse CXCL12 plasma levels, indicating in vivo interaction of the agonist with ACKR3. Whereas twice daily in vivo administration of the ACKR3 agonist lacked inhibitory effect on bleomycin-induced lung fibrosis, it had a modest, but significant anti-fibrotic effect in the carbon tetrachloride (CCl4)-induced liver fibrosis model. In the latter model, ACKR3 stimulation affected the expression of several fibrosis-related genes and led to reduced collagen content as determined by picro-sirius red staining and hydroxyproline quantification. These data confirm that ACKR3 agonism, at least to some extent, attenuates fibrosis, although this effect is rather modest and heterogeneous across various tissue types. Stimulating ACKR3 alone without intervening in other signaling pathways involved in the multicellular crosstalk leading to fibrosis will, therefore, most likely not be sufficient to deliver a satisfactory clinical outcome.
Collapse
Affiliation(s)
- Tom Van Loy
- Laboratory of Virology and Chemotherapy, Rega Institute, Department of Microbiology, Immunology and Transplantation, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Steven De Jonghe
- Laboratory of Virology and Chemotherapy, Rega Institute, Department of Microbiology, Immunology and Transplantation, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | | | - Wouter Dheedene
- Endothelial Cell Biology Unit, Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Reinout Stoop
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Lars Verschuren
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), Leiden, The Netherlands
| | | | - Patrick Chaltin
- CISTIM, Gaston Geenslaan 2, 3001, Leuven, Belgium
- Centre for Drug Design and Discovery (CD3), KU Leuven, Gaston Geenslaan 2, 3001, Leuven, Belgium
| | - Aernout Luttun
- Endothelial Cell Biology Unit, Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Dominique Schols
- Laboratory of Virology and Chemotherapy, Rega Institute, Department of Microbiology, Immunology and Transplantation, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| |
Collapse
|
10
|
Kondo R, Kawata N, Suzuki Y, Yamamura H. Ca<sup>2+</sup> Signaling and Proliferation <i>via</i> Ca<sup>2+</sup>-Sensing Receptors in Human Hepatic Stellate LX-2 Cells. Biol Pharm Bull 2022; 45:664-667. [DOI: 10.1248/bpb.b22-00103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Rubii Kondo
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Naoki Kawata
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Yoshiaki Suzuki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Hisao Yamamura
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| |
Collapse
|
11
|
Naltrexone protects against BDL-induced cirrhosis in Wistar rats by attenuating thrombospondin-1 and enhancing antioxidant defense system via Nrf-2. Life Sci 2022; 300:120576. [PMID: 35487305 DOI: 10.1016/j.lfs.2022.120576] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/17/2022] [Accepted: 04/20/2022] [Indexed: 11/23/2022]
Abstract
AIMS It is well-established that thrombospondin-1 (THBS-1), vascular endothelial growth factor-A (VEGF-A), nuclear factor-erythroid 2-related factor 2 (Nrf-2), Kelch-like ECH-associated protein 1 (Keap-1), and transforming growth factor-beta 1 (TGF-β1) are the pivotal players of liver fibrosis. Recent studies have shown that endogenous opioid levels increase during liver cirrhosis. Therefore, the present study aimed to clarify the effect of naltrexone (NTX), an opioid antagonist, on the alteration of these factors following bile duct ligation (BDL)-induced liver cirrhosis. MAIN METHODS Wistar male rats (n = 50) were categorized equally into 5 groups (baseline, sham+saline, BDL + saline, sham+NTX (10 mg/kg of body weight (BW)), and BDL + NTX (10 mg/kg of BW)). At the end of the experiment, H&E staining was used to assess necrosis and lobular damage of hepatic tissue. The gene expression of THBS-1 and NADPH oxidase 1 (NOX-1) was measured by real time-PCR and VEGF-A, Nrf-2, Keap-1, and TGF-β1 protein levels were assessed by western blot. The antioxidant enzymes activity, total oxidant status (TOS) and MDA level were measured by commercial kits. KEY FINDINGS Hepatic necrosis and lobular damage increased substantially and NTX reduced them markedly in the BDL group. Gene expression of hepatic THBS-1 and NOX-1, TOS and MDA levels increased markedly in the BDL + saline group, and Nrf-2 and VEGF-A values decreased significantly in the BDL + NTX group. NTX recovered THBS-1, NOX-1 and Nrf-2 in the BDL + NTX group, substantially (p-value ≤ 0.05). SIGNIFICANCE Data showed that NTX treatment attenuates liver fibrosis mainly by lowering THBS-1 and NOX-1 and increasing Nrf-2 protein level and antioxidant enzymes.
Collapse
|
12
|
Pi L, Yang L, Fang BR, Meng XX, Qian L. Exosomal microRNA-125a-3p from human adipose-derived mesenchymal stem cells promotes angiogenesis of wound healing through inhibiting PTEN. Mol Cell Biochem 2021; 477:115-127. [PMID: 34581942 DOI: 10.1007/s11010-021-04251-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/20/2021] [Indexed: 02/06/2023]
Abstract
Angiogenesis plays a key in the process of tissue repair and wound healing. Human adipose-derived mesenchymal stem cells (HADSCs) have been found to act a promotion role during angiogenesis. Moreover, miR-125a-3p in HADSCs could promote the angiogenesis of HUVECs, but their specific mechanism in wound healing needs further study. Western blotting and qRT-PCR were used for detecting the protein and mRNA level, respectively. Exosomes were isolated successfully, and transmission electron microscope was used to identify exosomes. Angiogenesis, cell migration, and proliferation were detected with tube formation, wound healing, and MTT assays. The interactions of miR-125a-3p and PTEN were validated using dual-luciferase reporter assay. Animal model was used to evaluate the effect of miR-125a-3p on wound healing. HADSCs-exosome remarkably promoted the viability, migration, and angiogenesis of HUVECs. Knockdown of miR-125a-3p in HADSCs could inhibit the effect of HADSCs-exosome, while overexpression of miR-125a-3p could further promote the effect of HADSCs-exosome on HUVECs. MiR-125a-3p from HADSCs-exosome inhibited the expression of PTEN in HUVECs. Knockdown of PTEN promoted the viability, migration, and angiogenesis of HUVECs and reversed the effect of miR-125a-3p knockdown on HUVECs. Finally, miR-125a-3p from HADSCs-exosome could promote wound healing and angiogenesis in mice by inhibiting PTEN in mice wound granulation tissues. MiR-125a-3p from the HADSCs-exosome promoted the wound healing and angiogenesis, and these effects were achieved through regulating PTEN. This study may provide a new thought for the treatment and prevention of tissue repair.
Collapse
Affiliation(s)
- Li Pi
- Department of Burn and Plastic Surgery, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Furong District, Changsha, 410011, Hunan Province, China
| | - Li Yang
- Department of Burn and Plastic Surgery, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Furong District, Changsha, 410011, Hunan Province, China
| | - Bai-Rong Fang
- Department of Burn and Plastic Surgery, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Furong District, Changsha, 410011, Hunan Province, China
| | - Xian-Xi Meng
- Department of Burn and Plastic Surgery, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Furong District, Changsha, 410011, Hunan Province, China
| | - Li Qian
- Department of Burn and Plastic Surgery, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Furong District, Changsha, 410011, Hunan Province, China.
| |
Collapse
|
13
|
Sun Y, Wang T, Wen QT, Yu DH, Chen JX. VEGF gene transfection restores the angiogenesis of oral submucous fibrosis in mice. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:930. [PMID: 34350245 PMCID: PMC8263869 DOI: 10.21037/atm-21-2213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/02/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND To explore the effectiveness of adenovirus-enhanced green fluorescent protein-vascular endothelial growth factor165 (AD-EGFP-VEGF165) transfection on fibroblasts from mice, and we assessed whether VEGF165 restores the angiogenesis of oral submucous fibrosis (OSF) in mice. METHODS AD-EGFP-VEGF165 and AD-EGFP were transfected into fibroblasts from mouse buccal tissues in vitro. The expression of VEGF before and after transfection was detected by RT-qPCR and ELISA in each group of fibroblasts. Fifteen OSF mice (pre-experimental construction) were randomly divided into 3 groups, and equal amounts of AD-EGFP-VEGF165 virus, AD-EGFP virus, and saline were injected into the buccal submucosal tissue of OSF mice. The expression of VEGF and local tissue angiogenesis were observed and measured in each group of animals. RESULTS The Ad-EGFP-VEGF165-transfected fibroblasts increased human and mouse VEGF expression compared to the Ad-EGFP group and control group (P<0.05). The buccal submucosal tissue of mice was injected with Ad-EGFP-VEGF165 after the 6th day, and the expression of VEGF was effectively expressed in AD-EGFP-VEGF165 group (P<0.05), while no positive expression observed in other groups. and the number of microvessels in the AD-EGFP-VEGF165 group increased significantly compared to the other groups (P<0.05). CONCLUSIONS Ad-EGFP-VEGF165 can be successfully transfected into fibroblasts from mice, and restored the angiogenesis of OSF in mice.
Collapse
Affiliation(s)
- Ying Sun
- Dental Medical Center, Hainan General Hospital (Hainan Affiliated Hospital of Hainan medical University), Haikou, China
| | - Tao Wang
- Dental Medical Center, Hainan General Hospital (Hainan Affiliated Hospital of Hainan medical University), Haikou, China
| | - Qi-Tao Wen
- Dental Medical Center, Hainan General Hospital (Hainan Affiliated Hospital of Hainan medical University), Haikou, China
| | - Da-Hai Yu
- Department of Stomatology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jing-Xin Chen
- Dental Medical Center, Hainan General Hospital (Hainan Affiliated Hospital of Hainan medical University), Haikou, China
| |
Collapse
|
14
|
Kiseleva YV, Zharikov YO, Maslennikov RV, Pavlov CS, Nikolenko VN. [Molecular factors associated with regression of liver fibrosis of alcoholic etiology]. TERAPEVT ARKH 2021; 93:204-208. [PMID: 36286638 DOI: 10.26442/00403660.2021.02.200617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023]
Abstract
Liver fibrosis develops as a result of chronic liver damage of various etiologies, is characterized by excessive synthesis of connective tissue by activated stellate liver cells. The toxic effect of alcohol is one of the most significant and common etiological factors worldwide. Stellate cell activation results from the interaction of multiple molecular fibrogenic pathways triggered by intracellular and extracellular, hepatic and extrahepatic stimuli. Data analysis showed that knowledge about these abnormal pathways and biomolecular processes may further contribute to the improvement of approaches to assessment of disease prognosis and treatment of alcoholic liver disease.
Collapse
Affiliation(s)
- Y V Kiseleva
- Sechenov First Moscow State Medical University (Sechenov University)
| | - Y O Zharikov
- Sechenov First Moscow State Medical University (Sechenov University)
| | - R V Maslennikov
- Sechenov First Moscow State Medical University (Sechenov University)
| | - C S Pavlov
- Sechenov First Moscow State Medical University (Sechenov University)
| | - V N Nikolenko
- Sechenov First Moscow State Medical University (Sechenov University)
- Lomonosov Moscow State University
| |
Collapse
|
15
|
Long-Term Aspartame Administration Leads to Fibrosis, Inflammasome Activation, and Gluconeogenesis Impairment in the Liver of Mice. BIOLOGY 2021; 10:biology10020082. [PMID: 33499218 PMCID: PMC7911935 DOI: 10.3390/biology10020082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Aspartame is an artificial sweetener used in foods and beverages worldwide. However, it is linked to oxidative stress, inflammation, and liver damage through mechanisms that are not fully elucidated yet. This work aimed to investigate the effects of long-term administration of aspartame on the oxidative and inflammatory mechanisms associated with liver fibrosis progression in mice. METHODS Mice were divided into two groups with six animals each: control and aspartame. Aspartame (80 mg/kg, via oral) or vehicle was administrated for 12 weeks. RESULTS Aspartame caused liver damage and elevated serum transaminase levels. Aspartame also generated liver fibrosis, as evidenced by histology analysis, and pro-fibrotic markers' upregulation, including transforming growth factor β 1, collagen type I alpha 1, and alpha-smooth muscle actin. Furthermore, aspartame reduced nuclear factor erythroid 2-related factor 2 (Nrf2) activation and enzymatic antioxidant activity and increased lipid peroxidation, which triggered NOD-like receptor containing protein 3 (NLRP3) inflammasome activation and p53 induction. Furthermore, aspartame reduced peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) levels, possibly through p53 activation. This PGC-1α deficiency could be responsible for the changes in lipid profile in serum, total lipid accumulation, and gluconeogenesis impairment in liver, evidenced by the gluconeogenic enzymes' downregulation, thus causing hypoglycemia. CONCLUSIONS This work provides new insights to understand the mechanisms related to the adverse effects of aspartame on liver tissue.
Collapse
|
16
|
Nerve Growth Factor (NGF) modulates in vitro induced myofibroblasts by highlighting a differential protein signature. Sci Rep 2021; 11:1672. [PMID: 33462282 PMCID: PMC7814037 DOI: 10.1038/s41598-021-81040-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 12/31/2020] [Indexed: 02/06/2023] Open
Abstract
We previously described the profibrogenic effect of NGF on conjunctival Fibroblasts (FBs) and its ability to trigger apoptosis in TGFβ1-induced myofibroblasts (myoFBs). Herein, cell apoptosis/signalling, cytokines' signature in conditioned media and inflammatory as well as angiogenic pathway were investigated. Experimental myoFBs were exposed to NGF (0.1-100 ng/mL), at defined time-point for confocal and biomolecular analysis. Cells were analysed for apoptotic and cell signalling activation in cell extracts and for some inflammatory and proinflammatory/angiogenic factors' activations. NGF triggered cJun overexpression and phospho-p65-NFkB nuclear translocation. A decreased Bcl2:Bax ratio and a significant expression of smad7 were confirmed in early AnnexinV-positive myoFBs. A specific protein signature characterised the conditioned media: a dose dependent decrease occurred for IL8, IL6 while a selective increase was observed for VEGF and cyr61 (protein/mRNA). TIMP1 levels were unaffected. Herein, NGF modulation of smad7, the specific IL8 and IL6 as well as VEGF and cyr61 modulation deserve more attention as opening to alternative approaches to counteract fibrosis.
Collapse
|
17
|
Yasmin A, Regan DP, Schook LB, Gaba RC, Schachtschneider KM. Transcriptional regulation of alcohol induced liver fibrosis in a translational porcine hepatocellular carcinoma model. Biochimie 2021; 182:73-84. [PMID: 33444661 DOI: 10.1016/j.biochi.2020.12.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/09/2020] [Accepted: 12/28/2020] [Indexed: 01/18/2023]
Abstract
Hepatocellular carcinoma (HCC) is the 5th most common and 2nd deadliest cancer worldwide. HCC risk factors include alcohol induced liver cirrhosis, which prompts hepatic inflammation, cell necrosis, and fibrosis deposition. As 25% of HCC cases are associated with alcohol induced liver disease, understanding the effects of the cirrhotic liver microenvironment on HCC tumor biology and therapeutic responses are critical. This study utilized the Oncopig Cancer Model-a transgenic pig model that recapitulates human HCC through induced expression of KRASG12D and TP53R167H driver mutations-to investigate the molecular mechanisms underlying alcohol induced liver disease. Oncopigs (n = 5) underwent fibrosis induction via infusion of ethanol and ethiodized oil (1:3 v/v dosed at 0.75 mL/kg) into the hepatic arterial circulation. Eight-weeks post induction, liver tissue samples from fibrotic and age-matched control (n = 5) Oncopigs were collected for histological evaluation and transcriptional profiling. Increased hepatic inflammation and fibrosis was observed in fibrotic Oncopigs via pathological assessment. Transcriptional profiling (RNA-seq) resulted in the identification of 4387 differentially expressed genes between Oncopig fibrotic and control livers. GO term enrichment analysis identified pathway alterations associated with cirrhosis progression in humans, including cell proliferation, angiogenesis, extracellular matrix deposition, and oxidation-reduction. Key alterations include activation of hepatic stellate cells, increased matrix metalloproteinase production, and altered expression of ABC and SLC transporter genes involved in transport of anticancer drugs.These results demonstrate Oncopig liver fibrosis recapitulates transcriptional hallmarks of human cirrhosis, making the Oncopig an ideal model for studying the effects of the cirrhotic liver microenvironment on HCC tumor biology and therapeutic response.
Collapse
Affiliation(s)
- Alvi Yasmin
- Department of Radiology, University of Illinois at Chicago, United States
| | - Daniel P Regan
- Flint Animal Cancer Center, Colorado State University, United States
| | - Lawrence B Schook
- Department of Radiology, University of Illinois at Chicago, United States; Department of Animal Sciences, University of Illinois at Urbana-Champaign, United States; National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, United States
| | - Ron C Gaba
- Department of Radiology, University of Illinois at Chicago, United States
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago, United States; National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, United States; Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, United States.
| |
Collapse
|
18
|
Jun JH, Jung J, Kim JY, Hwang SG, Bae SH, Kim GJ. Upregulation of C-Reactive Protein by Placenta-Derived Mesenchymal Stem Cells Promotes Angiogenesis in A Rat Model with Cirrhotic Liver. Int J Stem Cells 2020; 13:404-413. [PMID: 33122469 PMCID: PMC7691864 DOI: 10.15283/ijsc20052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 08/11/2020] [Accepted: 09/21/2020] [Indexed: 12/15/2022] Open
Abstract
Background and Objectives Liver cirrhosis is accompanied by abnormal vascular shunts. The Wnt pathway is essential for endothelial cell survival and proliferation. C-reactive protein (CRP), which is produced by hepatocyte, activates angiogenesis in cardiovascular diseases. Methods and Results The expression of CRP in CCl4-injured rat livers was detected using qRT-PCR and Western blotting after transplantation of placenta-derived mesenchymal stem cells (PD-MSCs) into rats. To determine whether CRP functions in hepatic regeneration by promoting angiogenesis through the Wnt pathway, we detected VEGF and β-catenin in liver tissues and BrdU and β-catenin in hepatocytes by immunofluorescence. The expression levels of CRP, Wnt pathway-related and angiogenic factors were increased in CCl4-injured and PD-MSCs transplanted rat livers. In vitro, the expression levels of Wnt signaling and angiogenic factors were decreased in siRNA-CRP-transfected rat hepatocytes. Conclusions CRP upregulation by PD-MSCs participates in vascular remodeling to promote liver regeneration via the Wnt signaling pathway during hepatic failure.
Collapse
Affiliation(s)
- Ji Hye Jun
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Jieun Jung
- Non-Clinical Evaluation Center, CHA Advanced Research Institute, Seongnam, Korea
| | - Jae Yeon Kim
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Seong-Gyu Hwang
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Si Hyun Bae
- Department of Internal Medicine, Catholic University Medical College, Seoul, Korea
| | - Gi Jin Kim
- Department of Biomedical Science, CHA University, Seongnam, Korea
| |
Collapse
|
19
|
Zadorozhna M, Di Gioia S, Conese M, Mangieri D. Neovascularization is a key feature of liver fibrosis progression: anti-angiogenesis as an innovative way of liver fibrosis treatment. Mol Biol Rep 2020; 47:2279-2288. [PMID: 32040707 DOI: 10.1007/s11033-020-05290-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 01/28/2020] [Indexed: 12/11/2022]
Abstract
Liver fibrosis affects over 100 million people in the world; it represents a multifactorial, fibro-inflammatory disorder characterized by exacerbated production of extracellular matrix with consequent aberration of hepatic tissue. The aetiology of this disease is very complex and seems to involve a broad spectrum of factors including the lifestyle, environment factors, genes and epigenetic changes. More evidences indicate that angiogenesis, a process consisting in the formation of new blood vessels from pre-existing vessels, plays a crucial role in the progression of liver fibrosis. Central to the pathogenesis of liver fibrosis is the hepatic stellate cells (HSCs) which represent a crossroad among inflammation, fibrosis and angiogenesis. Quiescent HSCs can be stimulated by a host of growth factors, pro-inflammatory mediators produced by damaged resident liver cell types, as well as by hypoxia, contributing to neoangiogenesis, which in turn can be a bridge between acute and chronic inflammation. As matter of fact, studies demonstrated that neutralization of vascular endothelial growth factor as well as other proangiogenic agents can attenuate the progression of liver fibrosis. With this review, our intent is to discuss the cause and the role of angiogenesis in liver fibrosis focusing on the current knowledge about the impact of anti-angiogenetic therapies in this pathology.
Collapse
Affiliation(s)
- Mariia Zadorozhna
- Department of Medical and Surgical Sciences, University of Foggia, Via Pinto 1, 71122, Foggia, Italy
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, Via Pinto 1, 71122, Foggia, Italy
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Via Pinto 1, 71122, Foggia, Italy
| | - Domenica Mangieri
- Department of Medical and Surgical Sciences, University of Foggia, Via Pinto 1, 71122, Foggia, Italy.
| |
Collapse
|
20
|
Coronary stents with inducible VEGF/HGF-secreting UCB-MSCs reduced restenosis and increased re-endothelialization in a swine model. Exp Mol Med 2018; 50:1-14. [PMID: 30174328 PMCID: PMC6119684 DOI: 10.1038/s12276-018-0143-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 05/30/2018] [Accepted: 06/07/2018] [Indexed: 12/12/2022] Open
Abstract
Atherosclerotic plaques within the vasculature may eventually lead to heart failure. Currently, cardiac stenting is the most effective and least invasive approach to treat this disease. However, in-stent restenosis is a complex chronic side effect of stenting treatment. This study used coronary stents coated with stem cells secreting angiogenic growth factors via an inducible genome-editing system to reduce stent restenosis and induce re-endothelialization within the artery. The characteristics of the cells and their adhesion properties on the stents were confirmed, and the stents were transplanted into a swine model to evaluate restenosis and the potential therapeutic use of stents with stem cells. Restenosis was evaluated using optical coherence tomography (OCT), microcomputed tomography (mCT) and angiography, and re-endothelialization was evaluated by immunostaining after cardiac stent treatment. Compared to a bare metal stent (BMS) or a parental umbilical cord blood-derived mesenchymal stem cell (UCB-MSC)-coated stent, the stents with stem cells capable of the controlled release of hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF) successfully reduced restenosis within the stent and induced natural re-endothelialization. Furthermore, UCB-MSCs exhibited the ability to differentiate into endothelial cells in Matrigel, and HGF and VEGF improved this differentiation. Our study indicates that stents coated with UCB-MSCs secreting VEGF/HGF reduce the restenosis side effects of cardiac stenting with improved re-endothelialization.
Collapse
|
21
|
Choudhari SS, Kulkarni DG, Patankar S, Kheur SM, Sarode SC, Sarode GS, Patil S. Angiogenesis and Fibrogenesis in Oral Submucous Fibrosis: A Viewpoint. J Contemp Dent Pract 2018; 19:242-245. [PMID: 29422478 DOI: 10.5005/jp-journals-10024-2244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Oral submucous fibrosis (OSF) is characterized by excessive fibrosis of submucosa. The degree of vascularity in OSF has always been a matter of debate. Angiogenesis is the key mechanism involved in regeneration and repair. It also plays an important role in various pathologic conditions. Angiogenesis may contribute to the progression of fibrosis in fibrotic disorders. Inhibition of pathological angiogenesis is considered to be a new strategy for the treatment of various fibrotic disorders. In OSF, angiogenesis can be related to progression fibrosis. This article briefly describes the role of angiogenesis in pathogenesis of fibrosis in OSF and the importance of inhibition of pathologic angiogenesis in its prevention and treatment. CLINICAL SIGNIFICANCE Understanding the association between angiogenesis and fibrogenesis can help in developing new therapeutic strategies for treatment of OSF.
Collapse
Affiliation(s)
- Sheetal S Choudhari
- Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College & Hospital, Dr. D. Y. Patil Vidyapeeth, Pune Maharashtra, India, Phone: +919819331220, e-mail:
| | - Deepak G Kulkarni
- Department of Oral and Maxillofacial Surgery, Dr. D. Y. Patil Dental College & Hospital, Dr. D. Y. Patil Vidyapeeth, Pune Maharashtra, India
| | - Sangeeta Patankar
- Department of Oral Pathology and Microbiology, Yerala Medical Trust Dental College & Research Institute, Navi Mumbai Maharashtra, India
| | - Supriya M Kheur
- Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College & Hospital, Dr. D. Y. Patil Vidyapeeth, Pune Maharashtra, India
| | - Sachin C Sarode
- Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College & Hospital, Dr. D. Y. Patil Vidyapeeth, Pune Maharashtra, India
| | - Gargi S Sarode
- Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College & Hospital, Dr. D. Y. Patil Vidyapeeth, Pune Maharashtra, India
| | - Shankargouda Patil
- Division of Oral Pathology, Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
22
|
Catani L, Sollazzo D, Bianchi E, Ciciarello M, Antoniani C, Foscoli L, Caraceni P, Giannone FA, Baldassarre M, Giordano R, Montemurro T, Montelatici E, D'Errico A, Andreone P, Giudice V, Curti A, Manfredini R, Lemoli RM. Molecular and functional characterization of CD133 + stem/progenitor cells infused in patients with end-stage liver disease reveals their interplay with stromal liver cells. Cytotherapy 2017; 19:1447-1461. [PMID: 28917627 DOI: 10.1016/j.jcyt.2017.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 08/01/2017] [Accepted: 08/03/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND AIMS Growing evidence supports the therapeutic potential of bone marrow (BM)-derived stem/progenitor cells for end-stage liver disease (ESLD). We recently demonstrated that CD133+ stem/progenitor cell (SPC) reinfusion in patients with ESLD is feasible and safe and improve, albeit transiently, liver function. However, the mechanism(s) through which BM-derived SPCs may improve liver function are not fully elucidated. METHODS Here, we characterized the circulating SPCs compartment of patients with ESLD undergoing CD133+ cell therapy. Next, we set up an in vitro model mimicking SPCs/liver microenvironment interaction by culturing granulocyte colony-stimulating factor (G-CSF)-mobilized CD133+and LX-2 hepatic stellate cells. RESULTS We found that patients with ESLD show normal basal levels of circulating hematopoietic and endothelial progenitors with impaired clonogenic ability. After G-CSF treatment, patients with ESLD were capable to mobilize significant numbers of functional multipotent SPCs, and interestingly, this was associated with increased levels of selected cytokines potentially facilitating SPC function. Co-culture experiments showed, at the molecular and functional levels, the bi-directional cross-talk between CD133+ SPCs and human hepatic stellate cells LX-2. Human hepatic stellate cells LX-2 showed reduced activation and fibrotic potential. In turn, hepatic stellate cells enhanced the proliferation and survival of CD133+ SPCs as well as their endothelial and hematopoietic function while promoting an anti-inflammatory profile. DISCUSSION We demonstrated that the interaction between CD133+ SPCs from patients with ESLD and hepatic stellate cells induces significant functional changes in both cellular types that may be instrumental for the improvement of liver function in cirrhotic patients undergoing cell therapy.
Collapse
Affiliation(s)
- Lucia Catani
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy.
| | - Daria Sollazzo
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy
| | - Elisa Bianchi
- Centre for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marilena Ciciarello
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy
| | - Chiara Antoniani
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy
| | - Licia Foscoli
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy
| | - Paolo Caraceni
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy; Center for Applied Biomedical Research (C.R.B.A.), Azienda Ospedaliero/Universitaria di Bologna, Bologna, Italy
| | | | - Maurizio Baldassarre
- Center for Applied Biomedical Research (C.R.B.A.), Azienda Ospedaliero/Universitaria di Bologna, Bologna, Italy
| | - Rosaria Giordano
- Cell Factory, Unit of Cellular Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Tiziana Montemurro
- Cell Factory, Unit of Cellular Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Elisa Montelatici
- Cell Factory, Unit of Cellular Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Antonia D'Errico
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Pietro Andreone
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Valeria Giudice
- Immunohematology Service and Blood Bank-Azienda Ospedaliero/Universitaria di Bologna, Bologna, Italy
| | - Antonio Curti
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy
| | - Rossella Manfredini
- Centre for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto Massimo Lemoli
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| |
Collapse
|
23
|
Cytokines, hepatic cell profiling and cell interactions during bone marrow cell therapy for liver fibrosis in cholestatic mice. PLoS One 2017; 12:e0187970. [PMID: 29176797 PMCID: PMC5703547 DOI: 10.1371/journal.pone.0187970] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 10/30/2017] [Indexed: 12/29/2022] Open
Abstract
Bone marrow cells (BMC) migrate to the injured liver after transplantation, contributing to regeneration through multiple pathways, but mechanisms involved are unclear. This work aimed to study BMC migration, characterize cytokine profile, cell populations and proliferation in mice with liver fibrosis transplanted with GFP+ BMC. Confocal microscopy analysis showed GFP+ BMC near regions expressing HGF and SDF-1 in the fibrotic liver. Impaired liver cell proliferation in fibrotic groups was restored after BMC transplantation. Regarding total cell populations, there was a significant reduction in CD68+ cells and increased Ly6G+ cells in transplanted fibrotic group. BMC contributed to the total populations of CD144, CD11b and Ly6G cells in the fibrotic liver, related to an increment of anti-fibrotic cytokines (IL-10, IL-13, IFN-γ and HGF) and reduction of pro-inflammatory cytokines (IL-17A and IL-6). Therefore, HGF and SDF-1 may represent important chemoattractants for transplanted BMC in the injured liver, where these cells can give rise to populations of extrahepatic macrophages, neutrophils and endothelial progenitor cells that can interact synergistically with other liver cells towards the modulation of an anti-fibrotic cytokine profile promoting the onset of liver regeneration.
Collapse
|
24
|
Alizai PH, Bertram L, Kroy D, Kummer J, Andert A, Neumann UP, Ulmer TF, Fragoulis A. Expression of VEGFR-2 during Liver Regeneration after Partial Hepatectomy in a Bioluminescence Mouse Model. Eur Surg Res 2017; 58:330-340. [PMID: 29073598 DOI: 10.1159/000479628] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Liver regeneration requires the formation of new blood vessels. Endothelial cell proliferation is stimulated by vascular endothelial growth factor (VEGF) and its receptor tyrosine kinase VEGFR-2. The aim of this study was to investigate VEGFR-2 expression in vivo during liver regeneration after partial hepatectomy (PHx). METHODS Transgenic VEGFR-2-luc mice were used in which the luciferase reporter gene was under control of the VEGFR-2 promoter. Following 2/3 PHx, the mice underwent in vivo bioluminescence imaging until the 14th postoperative day. Additionally, liver tissue was analyzed by immunohistochemistry, in vitro luminescence assays, and quantitative RT-PCR. RESULTS In vivo bioluminescence imaging showed a significant increase in VEGFR-2 promoter activity after PHx. Maximum signal was recorded on the 3rd day; 8 days postoperatively the signal intensity decreased significantly. On the 14th day, bioluminescence signal reached almost baseline levels. Immunohistochemistry, quantitative RT-PCR, and in vitro luminescence confirmed a significant increase on the 3rd day following resection. The mRNA expression of VEGFR-2 was significantly higher on day 3 than preoperatively as well as on day 8. CONCLUSION In vivo bioluminescence imaging with transgenic VEGFR-2-luc mice is feasible and provides a convenient model for noninvasively studying VEGFR-2 expression during liver regeneration. This may facilitate further experiments with modulation of angiogenesis by different substances.
Collapse
Affiliation(s)
- Patrick Hamid Alizai
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Lea Bertram
- Department of Surgery, Luisenhospital Aachen, Aachen, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University Hospital, Aachen, Germany
| | - Daniela Kroy
- Department of Gastroenterology and Metabolic Disorders, RWTH Aachen University Hospital, Aachen, Germany
| | - Julia Kummer
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany.,Department of Gynaecology and Obstetrics, Vivantes Clinic Berlin, Berlin, Germany
| | - Anne Andert
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Ulf Peter Neumann
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Tom Florian Ulmer
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Athanassious Fragoulis
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
25
|
Wang L, Zhao X, Feng Y, Ma X, Wu H, Zhu Q. Intrahepatic angiogenesis increases portal hypertension in hepatitis B patients with cirrhosis. Hepatol Res 2017; 47:E94-E103. [PMID: 27115574 DOI: 10.1111/hepr.12732] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/19/2016] [Accepted: 04/22/2016] [Indexed: 01/17/2023]
Abstract
AIM It remains unclear whether intrahepatic angiogenesis increases portal hypertension (PH) in hepatitis B with cirrhosis. We aim to investigate the relationship between intrahepatic angiogenesis and PH in hepatitis B patients with cirrhosis. METHODS Sixty hepatitis B patients with cirrhosis and 40 healthy subjects were included in this study. Angiogenesis markers vascular endothelial growth factor receptor-2 (VEGFR2), von Willebrand factor (vWF), and fibrosis marker α-smooth muscle actin (α-SMA) were observed by immunohistochemistry. Sirius Red staining was also used to determine liver fibrosis. Correlations between levels of intrahepatic angiogenesis and Child-Pugh classes, liver fibrosis degree, and portal vein pressure were examined. We also analyzed the relationship between levels of intrahepatic angiogenesis and complications of PH, including esophageal varices (EV), ascites, and hypersplenism. RESULTS Correlation was observed between the levels of VEGFR2 (r = 0.590, P < 0.01), vWF (r = 0.524, P < 0.01) in tissue, and Child-Pugh classes. Significant correlations were observed between levels of VEGFR2 and α-SMA (r = 0.710, P < 0.01), VEGFR2 and Sirius Red (r = 0.841, P < 0.01), vWF and α-SMA (r = 0.768, P < 0.01), and vWF and Sirius Red (r = 0.825, P < 0.01). Patients with hepatic venous pressure gradient (HVPG) ≥12 mmHg showed higher levels of VEGFR2 and vWF expression compared to those with (HVPG) <12 mmHg (2.60 ± 1.28% vs. 1.09 ± 0.73%; 5.85 ± 2.45% vs. 2.31 ± 1.34%, P < 0.01), respectively. Moreover, complications of PH, including size of esophageal varices (P < 0.01), presence of ascites (P < 0.01), and spleen volume (P < 0.01) were significantly affected by the levels of intrahepatic angiogenesis. CONCLUSION Intrahepatic angiogenesis increases PH in hepatitis B patients with cirrhosis. The study provides the potential ways to intervene in the progresses for therapeutic benefits in cirrhosis and PH.
Collapse
Affiliation(s)
- Le Wang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Xinya Zhao
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yuemin Feng
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Xiaowen Ma
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Hao Wu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Qiang Zhu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| |
Collapse
|
26
|
Arias N, Méndez M, Alcalde I, Íñigo-Portugués A, Merayo-Lloves J, Arias J, Arias JL. Assessing the brain through the eye: New ways to explore hepatic encephalopathy. Physiol Behav 2017; 173:263-271. [PMID: 28238775 DOI: 10.1016/j.physbeh.2017.02.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/21/2017] [Accepted: 02/21/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Minimal hepatic encephalopathy (mHE) has been shown to affect daily functioning, quality of life, driving and overall mortality. However, little is known about treating or diagnosing early impairments in mHE. We studied one of its precipitating factors, portal hypertension which is driving the inflammatory process behind mHE. The purpose was to describe an indirect diagnostic method able to detect the pathology at early stages based on the study of the vascularization and mast cells conjunctival hyperplasia as secondary inflammatory response associated to portal hypertension. Finally, we correlated the presence of histological changes in the eye in mHE with deficits in behavioral task acquisition. METHODS Rats were trained on a stimulus-response task and a spatial working memory task using the Morris water maze. Two groups of animals were used: a SHAM (sham-operated) group (n=10) and a portal hypertension (HT) group (n=10). The triple portal vein ligation method was used to create an animal model of mHE. Latencies to reach the platform, number of glial fibrillary acidic protein-immunoreactive astrocytes (GFAP-IR), mast cell expression and presence/absence of blood and lymphatic vessels were examined. RESULTS There were differences in stimulus-response behavioral performance, with a deficit in the acquisition in the HT group. However, no differences between groups were found on the spatial working memory task. At the same time, differences between groups were found in the GFAP-IR density, which was lower in the HT group, and in the number of mast cells and the presence of vessels, which were higher in the HT group. CONCLUSIONS In this study, we provide the first preliminary insight into the validity of exploring the eye as a possible tool to assess the diagnosis of mHE conditions.
Collapse
Affiliation(s)
- Natalia Arias
- Institute for Liver and Digestive Health, University College of London, Department of Medicine, Rowland Hill Street, London, UK; INEUROPA, Instituto de Neurociencias del Principado de Asturias, Spain.
| | - Marta Méndez
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Spain; Laboratorio de Neurociencias, Departamento de Psicología, Universidad de Oviedo, Plaza Feijoo s/n, 33003 Oviedo, Spain
| | - Ignacio Alcalde
- Fundación de Investigación Oftalmológica (FIO), Instituto Oftalmológico Fernández-Vega, Avda. Dres Fernández-Vega, 34, 33012 Oviedo, Spain
| | - Almudena Íñigo-Portugués
- Fundación de Investigación Oftalmológica (FIO), Instituto Oftalmológico Fernández-Vega, Avda. Dres Fernández-Vega, 34, 33012 Oviedo, Spain
| | - Jesús Merayo-Lloves
- Fundación de Investigación Oftalmológica (FIO), Instituto Oftalmológico Fernández-Vega, Avda. Dres Fernández-Vega, 34, 33012 Oviedo, Spain
| | - Jaime Arias
- Surgery Department, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Jorge L Arias
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Spain; Laboratorio de Neurociencias, Departamento de Psicología, Universidad de Oviedo, Plaza Feijoo s/n, 33003 Oviedo, Spain
| |
Collapse
|
27
|
Li Y, Turpin CP, Wang S. Role of thrombospondin 1 in liver diseases. Hepatol Res 2017; 47:186-193. [PMID: 27492250 PMCID: PMC5292098 DOI: 10.1111/hepr.12787] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/13/2016] [Accepted: 08/01/2016] [Indexed: 02/06/2023]
Abstract
Thrombospondin 1 (TSP1) is a matricellular glycoprotein that can be secreted by many cell types. Through binding to extracellular proteins and/or cell surface receptors, TSP1 modulates a variety of cellular functions. Since its discovery in 1971, TSP1 has been found to play important roles in multiple biological processes including angiogenesis, apoptosis, latent transforming growth factor-β activation, and immune regulation. Thrombospondin 1 is also involved in regulating many organ functions. However, the role of TSP1 in liver diseases has not been extensively addressed. In this review, we summarize the findings about the possible role that TSP1 plays in chronic liver diseases focusing on non-alcoholic fatty liver diseases, liver fibrosis, and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yanzhang Li
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
- Medical College of Henan University, Kaifeng, Henan 475004, China
| | - Courtney P Turpin
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
28
|
Hong Y, Han YQ, Wang YZ, Gao JR, Li YX, Liu Q, Xia LZ. Paridis Rhizoma Sapoinins attenuates liver fibrosis in rats by regulating the expression of RASAL1/ERK1/2 signal pathway. JOURNAL OF ETHNOPHARMACOLOGY 2016; 192:114-122. [PMID: 27396351 DOI: 10.1016/j.jep.2016.07.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 05/10/2016] [Accepted: 07/06/2016] [Indexed: 06/06/2023]
Abstract
ETHNO-PHARMACOLOGICAL RELEVANCE Paridis Rhizoma is a Chinese medicinal herb that has been used in liver disease treatment for thousands of years. Our previous studies found that Paridis Rhizoma saponins (PRS) are the critical components of Paridis Rhizoma which has good liver protection effect. However, the anti-hepatic fibrosis effect and the mechanism of PRS have seldom been reported. AIM OF THE STUDY To investigate the potential of PRS in the treatment of experimental liver fibrosis and the underlying mechanism. MATERIALS AND METHODS The chemical feature fingerprint of PRS was analyzed by UPLC-PDA. A total of 40 Male Sprague-Dawley (SD) rats were randomly divided into the control group, the model group, the PRS high dose group (PRS H) and the PRS low dose group (PRS L) with 10 rats in each group. The model, PRS H and L groups as liver fibrosis models were established with carbon tetrachloride (CCl4) method. PRS H and L groups were adopted PRS (300 and 150mg/kgd-1) treatment since the twelfth week of modeling till the sixteenth week. Pathological changes in hepatic tissue were examined using hematoxylin and eosin (H&E) and MASSON trichrome staining. Immunohistochemical analysis was performed to determine the protein expression of the RASAL1. RT-PCR and western blotting were used to detect the expression of ERK1/2 mRNA and protein. RESULTS Four saponins in PRS were identified from 19 detected chromatographic peaks on UPLC-PDA by comparing to the standard compounds. PRS can improve the degeneration and necrosis of hepatic tissue, reduce the extent of its fibrous hyperplasia according to H&E and MASSON staining detection. As was detected in PRS H and L groups, PRS down-regulated p-ERK1/2 mRNA and RASAL1 protein, and up-regulated the level of p-ERK1/2 mRNA and RASAL1 protein. CONCLUSION These results demonstrated that PRS can attenuate CCl4-induced liver fibrosis through the regulation of RAS/ERK1/2 signal pathway.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Carbon Tetrachloride
- Chemical and Drug Induced Liver Injury/enzymology
- Chemical and Drug Induced Liver Injury/pathology
- Chemical and Drug Induced Liver Injury/prevention & control
- Chromatography, High Pressure Liquid
- Cytoprotection
- GTPase-Activating Proteins/genetics
- GTPase-Activating Proteins/metabolism
- Gene Expression Regulation, Enzymologic
- Hyperplasia
- Immunohistochemistry
- Liver/drug effects
- Liver/enzymology
- Liver/pathology
- Liver Cirrhosis, Experimental/chemically induced
- Liver Cirrhosis, Experimental/enzymology
- Liver Cirrhosis, Experimental/pathology
- Liver Cirrhosis, Experimental/prevention & control
- Male
- Melanthiaceae/chemistry
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/genetics
- Mitogen-Activated Protein Kinase 3/metabolism
- Necrosis
- Phosphorylation
- Phytotherapy
- Plant Extracts/isolation & purification
- Plant Extracts/pharmacology
- Plants, Medicinal
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Reverse Transcriptase Polymerase Chain Reaction
- Saponins/isolation & purification
- Saponins/pharmacology
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Yan Hong
- Anhui University of Chinese Medicine, Hefei, Anhui 230031, China.
| | - Yan-Quan Han
- The First Affiliated Hospital, Anhui University of Chinese Medicine, Grade 3 Laboratory of TCM Preparation, State Administration of Anhui University of Chinese Medicine, Hefei, Anhui 230031, China.
| | - Yong-Zhong Wang
- The First Affiliated Hospital, Anhui University of Chinese Medicine, Grade 3 Laboratory of TCM Preparation, State Administration of Anhui University of Chinese Medicine, Hefei, Anhui 230031, China.
| | - Jia-Rong Gao
- The First Affiliated Hospital, Anhui University of Chinese Medicine, Grade 3 Laboratory of TCM Preparation, State Administration of Anhui University of Chinese Medicine, Hefei, Anhui 230031, China.
| | - Yu-Xin Li
- Anhui University of Chinese Medicine, Hefei, Anhui 230031, China; The First Affiliated Hospital, Anhui University of Chinese Medicine, Grade 3 Laboratory of TCM Preparation, State Administration of Anhui University of Chinese Medicine, Hefei, Anhui 230031, China.
| | - Qing Liu
- Anhui University of Chinese Medicine, Hefei, Anhui 230031, China; The First Affiliated Hospital, Anhui University of Chinese Medicine, Grade 3 Laboratory of TCM Preparation, State Administration of Anhui University of Chinese Medicine, Hefei, Anhui 230031, China.
| | - Lun-Zhu Xia
- The First Affiliated Hospital, Anhui University of Chinese Medicine, Grade 3 Laboratory of TCM Preparation, State Administration of Anhui University of Chinese Medicine, Hefei, Anhui 230031, China.
| |
Collapse
|
29
|
Remodeling of the Extracellular Matrix by Endothelial Cell-Targeting siRNA Improves the EPR-Based Delivery of 100 nm Particles. Mol Ther 2016; 24:2090-2099. [PMID: 27633444 DOI: 10.1038/mt.2016.178] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/07/2016] [Indexed: 01/22/2023] Open
Abstract
A number of nano drug delivery systems have recently been developed for cancer treatment, most of which are based on the enhanced permeability and retention effect. The advantages of the enhanced permeability and retention effect can be attributed to immature vasculature. Herein we evaluated the intratumoral distribution of lipid nanoparticles when the VEGF receptor 2 on tumor endothelial cells was inhibited by liposomal siRNA. VEGF receptor 2 inhibition resulted in an increase in intratumoral distribution and therapeutic efficacy despite the maturation of the tumor vasculature. A small molecule inhibitor against matrix metalloproteinase and macrophage depletion cancelled the improvement in the distribution of the lipid nanoparticles, suggesting that remodeling of tumor microenvironment played a role in the facilitated intratumoral distribution via the down-regulation of VEGF receptor 2. Accordingly, our results suggest that the enhanced permeability and retention effect is dependent, not only on the structure of the tumor vasculature, but also on the dynamics of the tumor microenvironment including extracellular matrix remodeling. Regulating the tumor microenvironment and the extracellular matrix by delivering tumor endothelial cell-targeting siRNA could potentiate the enhanced permeability and retention effect-based strategy.
Collapse
|
30
|
The Effects of Taoren-Honghua Herb Pair on Pathological Microvessel and Angiogenesis-Associated Signaling Pathway in Mice Model of CCl4-Induced Chronic Liver Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:2974256. [PMID: 27293456 PMCID: PMC4879235 DOI: 10.1155/2016/2974256] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/18/2016] [Indexed: 12/17/2022]
Abstract
Chronic liver disease is one of the most common diseases that threaten human health. Effective treatment is still lacking in western medicine. Semen Persicae (Taoren) and Flos Carthami (Honghua) are known to relieve acute hepatic injury and inflammation, improve microcirculation, and reduce tissue fiber. The aim of our study is to investigate the potential mechanisms of Taoren-Honghua Herb Pair (THHP) in murine model of chronic liver disease caused by Carbon Tetrachloride (CCl4). Mice were randomly divided into seven groups: (1) blank, (2) model, (3) control (colchicine, 0.1 mg/kg), (4) THHP (5.53, 2.67, and 1.33 g/kg), and (5) Tao Hong Siwu Decoction (THSWD) (8.50 g/kg). Histological change and microvessels density were examined by microscopy. Hepatic function, serum fibrosis related factors, and hepatic vascular endothelial growth factor (VEGF) were measured with ELISA. VEGF, kinase insert domain-containing receptor (KDR), Flt-1, and Akt mRNA expression in hepatic tissue were determined with PCR. Tissues of Akt, pAkt, KDR, and Flt-1 were measured with western blotting. Data from this study showed that THHP improved hepatic function and restrained the hepatic inflammation and fibrosis. Its role in inhibiting pathological angiogenesis and hepatic fibrogenesis may be through affecting the angiogenesis-associated VEGF and its upstream and downstream signaling pathways.
Collapse
|
31
|
Hellerbrand C, Massoumi R. Cylindromatosis-A Protective Molecule against Liver Diseases. Med Res Rev 2016; 36:342-59. [DOI: 10.1002/med.21381] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 11/12/2015] [Accepted: 11/17/2015] [Indexed: 12/27/2022]
Affiliation(s)
- Claus Hellerbrand
- Department of Internal Medicine I; University Hospital Regensburg; 93053 Regensburg Germany
| | - Ramin Massoumi
- Department of Laboratory Medicine, Medicon Village; Lund University; 22381 Lund Sweden
| |
Collapse
|