1
|
Nair B, Kamath AJ, Tergaonkar V, Sethi G, Nath LR. Mast cells and the gut-liver Axis: Implications for liver disease progression and therapy. Life Sci 2024; 351:122818. [PMID: 38866220 DOI: 10.1016/j.lfs.2024.122818] [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: 03/22/2024] [Revised: 05/24/2024] [Accepted: 06/07/2024] [Indexed: 06/14/2024]
Abstract
The role of mast cells, traditionally recognized for their involvement in immediate hypersensitivity reactions, has garnered significant attention in liver diseases. Studies have indicated a notable increase in mast cell counts following hepatic injury, underscoring their potential contribution to liver disorder pathogenesis. Predominantly situated in connective tissue that envelops the hepatic veins, bile ducts, and arteries, mast cells are central to both initiating and perpetuating liver disorders. Additionally, they are crucial for maintaining gastrointestinal barrier function. The gut-liver axis emphasizes the complex, two-way communication between the gut microbiome and the liver. Past research has implicated gut microbiota and their metabolites in the progression of hepatic disorders. This review sheds light on how mast cells are activated in various liver conditions such as alcoholic liver disease (ALD), non-alcoholic fatty liver disease (NAFLD), viral hepatitis, hepatic fibrogenesis, and hepatocellular carcinoma. It also briefly explores the connection between the gut microbiome and mast cell activation in these hepatic conditions.
Collapse
Affiliation(s)
- Bhagyalakshmi Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala 682041, India; Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala 682041, India
| | - Adithya Jayaprakash Kamath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala 682041, India; Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala 682041, India
| | - Vinay Tergaonkar
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore.
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala 682041, India.
| |
Collapse
|
2
|
Hardesty J, Hawthorne M, Day L, Warner J, Warner D, Gritsenko M, Asghar A, Stolz A, Morgan T, McClain C, Jacobs J, Kirpich IA. Steroid responsiveness in alcohol-associated hepatitis is linked to glucocorticoid metabolism, mitochondrial repair, and heat shock proteins. Hepatol Commun 2024; 8:e0393. [PMID: 38437061 PMCID: PMC10914234 DOI: 10.1097/hc9.0000000000000393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/15/2023] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Alcohol-associated hepatitis (AH) is one of the clinical presentations of alcohol-associated liver disease. AH has poor prognosis, and corticosteroids remain the mainstay of drug therapy. However, ~40% of patients do not respond to this treatment, and the mechanisms underlying the altered response to corticosteroids are not understood. The current study aimed to identify changes in hepatic protein expression associated with responsiveness to corticosteroids and prognosis in patients with AH. METHODS Patients with AH were enrolled based on the National Institute on Alcohol Abuse and Alcoholism inclusion criteria for acute AH and further confirmed by a diagnostic liver biopsy. Proteomic analysis was conducted on liver samples acquired from patients with AH grouped as nonresponders (AH-NR, n = 7) and responders (AH-R, n = 14) to corticosteroids, and nonalcohol-associated liver disease controls (n = 10). The definition of responders was based on the clinical prognostic model, the Lille Score, where a score < 0.45 classified patients as AH-R and a score > 0.45 as AH-NR. Primary outcomes used to assess steroid response were Lille Score (eg, improved liver function) and survival at 24 weeks. RESULTS Reduced levels of the glucocorticoid receptor and its transcriptional co-activator, glucocorticoid modulatory element-binding protein 2, were observed in the hepatic proteome of AH-NR versus AH-R. The corticosteroid metabolizing enzyme, 11-beta-hydroxysteroid dehydrogenase 1, was increased in AH-NR versus AH-R along with elevated mitochondrial DNA repair enzymes, while several proteins of the heat shock pathway were reduced. Analysis of differentially expressed proteins in AH-NR who survived 24 weeks relative to AH-NR nonsurvivors revealed several protein expression changes, including increased levels of acute phase proteins, elevated coagulation factors, and reduced mast cell markers. CONCLUSIONS This study identified hepatic proteomic changes that may predict responsiveness to corticosteroids and mortality in patients with AH.
Collapse
Affiliation(s)
- Josiah Hardesty
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Meghan Hawthorne
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, Kentucky, USA
| | - Le Day
- Department of Biological Sciences, Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Jeffrey Warner
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Dennis Warner
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, Kentucky, USA
| | - Marina Gritsenko
- Department of Biological Sciences, Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Aliya Asghar
- Department of Medicine and Research Services, Medicine and Research Services, VA Long Beach Healthcare System, Long Beach, California, USA
| | - Andrew Stolz
- Department of Medicine, Division of Gastrointestinal and Liver Disease, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Timothy Morgan
- Department of Medicine and Research Services, Medicine and Research Services, VA Long Beach Healthcare System, Long Beach, California, USA
| | - Craig McClain
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Robley Rex Veterans Medical Center, Louisville, Kentucky, USA
- Department of Medicine, University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Department of Medicine, University of Louisville Hepatobiology and Toxicology Center, University of Louisville School of Medicine, Louisville, Kentucky USA
| | - Jon Jacobs
- Department of Biological Sciences, Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Irina A. Kirpich
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Department of Medicine, University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Department of Medicine, University of Louisville Hepatobiology and Toxicology Center, University of Louisville School of Medicine, Louisville, Kentucky USA
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
3
|
Shu L, Li X, Liu Z, Li K, Shi A, Tang Y, Zhao L, Huang L, Zhang Z, Zhang D, Huang S, Lian S, Sheng G, Yan Z, Zhang Z, Xu Y. Bile exosomal miR-182/183-5p increases cholangiocarcinoma stemness and progression by targeting HPGD and increasing PGE2 generation. Hepatology 2024; 79:307-322. [PMID: 37140231 DOI: 10.1097/hep.0000000000000437] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/22/2023] [Indexed: 05/05/2023]
Abstract
BACKGROUND AIMS Cholangiocarcinoma (CCA) is a highly lethal malignancy originating from the biliary ducts. Current CCA diagnostic and prognostic assessments cannot satisfy the clinical requirement. Bile detection is rarely performed, and herein, we aim to estimate the clinical significance of bile liquid biopsy by assessing bile exosomal concentrations and components. APPROACH RESULTS Exosomes in bile and sera from CCA, pancreatic cancer, and common bile duct stone were identified and quantified by transmission electronmicroscopy, nanoparticle tracking analysis, and nanoFCM. Exosomal components were assessed by liquid chromatography with tandem mass spectrometry and microRNA sequencing (miRNA-seq). Bile exosomal concentration in different diseases had no significant difference, but miR-182-5p and miR-183-5p were ectopically upregulated in CCA bile exosomes. High miR-182/183-5p in both CCA tissues and bile indicates a poor prognosis. Bile exosomal miR-182/183-5p is secreted by CCA cells and can be absorbed by biliary epithelium or CCA cells. With xenografts in humanized mice, we showed that bile exosomal miR-182/183-5p promotes CCA proliferation, invasion, and epithelial-mesenchymal transition (EMT) by targeting hydroxyprostaglandin dehydrogenase in CCA cells and mast cells (MCs), and increasing prostaglandin E2 generation, which stimulates PTGER1 and increases CCA stemness. In single-cell mRNA-seq, hydroxyprostaglandin dehydrogenase is predominantly expressed in MCs. miR-182/183-5p prompts MC to release VEGF-A release from MC by increasing VEGF-A expression, which facilitates angiogenesis. CONCLUSIONS CCA cells secret exosomal miR-182/183-5p into bile, which targets hydroxyprostaglandin dehydrogenase in CCA cells and MCs and increases prostaglandin E2 and VEGF-A release. Prostaglandin E2 promotes stemness by activating PTGER1. Our results reveal a type of CCA self-driven progression dependent on bile exosomal miR-182/183-5p and MCs, which is a new interplay pattern of CCA and bile.
Collapse
Affiliation(s)
- Lizhuang Shu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xingyong Li
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Hepatobiliary Surgery, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zengli Liu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of General Surgery, Qilu Hospital (Qingdao), Shandong University, Qingdao, Shandong, China
| | - Kangshuai Li
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Anda Shi
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yongchang Tang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Liming Zhao
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lingling Huang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology(Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhiyue Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology(Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Dong Zhang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Shaohui Huang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Shuo Lian
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Guoli Sheng
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhangdi Yan
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zongli Zhang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yunfei Xu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
4
|
Kong L, Domarecka E, Szczepek AJ. Histamine and Its Receptors in the Mammalian Inner Ear: A Scoping Review. Brain Sci 2023; 13:1101. [PMID: 37509031 PMCID: PMC10376984 DOI: 10.3390/brainsci13071101] [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: 06/28/2023] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Histamine is a widely distributed biogenic amine with multiple biological functions mediated by specific receptors that determine the local effects of histamine. This review aims to summarize the published findings on the expression and functional roles of histamine receptors in the inner ear and to identify potential research hotspots and gaps. METHODS A search of the electronic databases PubMed, Web of Science, and OVID EMBASE was performed using the keywords histamine, cochlea*, and inner ear. Of the 181 studies identified, 18 eligible publications were included in the full-text analysis. RESULTS All four types of histamine receptors were identified in the mammalian inner ear. The functional studies of histamine in the inner ear were mainly in vitro. Clinical evidence suggests that histamine and its receptors may play a role in Ménière's disease, but the exact mechanism is not fully understood. The effects of histamine on hearing development remain unclear. CONCLUSIONS Existing studies have successfully determined the expression of all four histamine receptors in the mammalian inner ear. However, further functional studies are needed to explore the potential of histamine receptors as targets for the treatment of hearing and balance disorders.
Collapse
Affiliation(s)
- Lingyi Kong
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Ewa Domarecka
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Agnieszka J Szczepek
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Faculty of Medicine and Health Sciences, University of Zielona Gora, 65-046 Zielona Gora, Poland
| |
Collapse
|
5
|
Yurdakul Ertan H, Özbakış Dengiz G, Barut F, Yılmaz Ürün Y, Can M, Umer Hussen S, Üstündağ Y, Ustundag Y. The Investigation of Therapeutic Implications of Mast Cell Stabilizer Cromolyn Sodium on Cholestasis and Cholestatic Pruritus in Experimental Cholestasis. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2023; 34:62-72. [PMID: 36098363 PMCID: PMC9984916 DOI: 10.5152/tjg.2022.21744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Relevant studies have indicated that hepatic mast cells may have potential roles in the progression of cholestasis and cholestasis-induced itch. We aimed to compare the effects of cromolyn sodium and other medications on cholestatic pruritus, serum biochemistry, histamine, total bile acids, autotaxin, liver histopathology, and mast cell distribution in tissues in an experimental cholestasis model conducted by bile duct ligation. METHODS Rats received the determined treatment consecutively for 10 days in addition to bile duct ligation. On the 5th and 10th days of the experiment, the rats' itching behaviors were observed for 5 minutes. After 10 days, blood and tissue samples were taken. RESULTS Significant decreases in serum histamine and autotaxin levels, plasma total bile acids, total bilirubin, and biliary enzymes were reported only in cromolyn sodium-treated rats compared to the control group. In immunohistochemistry of the liver samples, the peribiliary mast cells stained positive for autotaxin. Except for bile duct infarctus, all histopathological findings of cholestasis significantly improved only in cromolyn sodium-treated and sertraline-treated rats. The liver and peritoneal mast cells significantly decreased only in cromolyn sodium-treated rats compared to the control group. On the 10th day of the experiment, the mean duration of itching was significantly lower in all groups, except for naloxone- and ondansetron-treated rats. CONCLUSION Cromolyn sodium has promising antipruritic efficacy and provides biochemical and histopathological recovery of the relevant parameters of cholestasis induced by bile duct ligation. For the first time in the literature, we showed that peribiliary mast cells can produce autotaxin, which is a very important pruritogenic signal in the setting of cholestasis.
Collapse
Affiliation(s)
- Hanife Yurdakul Ertan
- Department of Internal Medicine, Zonguldak Bülent Ecevit University Faculty of Medicine, Zonguldak, Turkey
| | - Günnur Özbakış Dengiz
- Department of Medical Pharmacology, Zonguldak Bülent Ecevit University Faculty of Medicine, Zonguldak, Turkey
| | - Figen Barut
- Department of Pathology, Zonguldak Bülent Ecevit University Faculty of Medicine, Zonguldak, Turkey
| | - Yonca Yılmaz Ürün
- Department of Gastroenterology and Hepatology, Eskişehir City Hospital, Turkey,Corresponding author: Yonca Yılmaz Ürün, e-mail:
| | - Murat Can
- Department of Medical Biochemistry, Zonguldak Bülent Ecevit University Faculty of Medicine, Zonguldak, Turkey
| | - Shemsu Umer Hussen
- Department of Medical Pharmacology, Zonguldak Bülent Ecevit University Faculty of Medicine, Zonguldak, Turkey
| | - Yücel Üstündağ
- Department of Gastroenterology and Hepatology, Zonguldak Bülent Ecevit University Faculty of Medicine, Zonguldak, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Huang S, Wu H, Luo F, Zhang B, Li T, Yang Z, Ren B, Yin W, Wu D, Tai S. Exploring the role of mast cells in the progression of liver disease. Front Physiol 2022; 13:964887. [PMID: 36176778 PMCID: PMC9513450 DOI: 10.3389/fphys.2022.964887] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/28/2022] [Indexed: 11/22/2022] Open
Abstract
In addition to being associated with allergic diseases, parasites, bacteria, and venoms, a growing body of research indicates that mast cells and their mediators can regulate liver disease progression. When mast cells are activated, they degranulate and release many mediators, such as histamine, tryptase, chymase, transforming growth factor-β1 (TGF-β1), tumor necrosis factor–α(TNF-α), interleukins cytokines, and other substances that mediate the progression of liver disease. This article reviews the role of mast cells and their secretory mediators in developing hepatitis, cirrhosis and hepatocellular carcinoma (HCC) and their essential role in immunotherapy. Targeting MC infiltration may be a novel therapeutic option for improving liver disease progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dehai Wu
- *Correspondence: Sheng Tai, ; Dehai Wu,
| | - Sheng Tai
- *Correspondence: Sheng Tai, ; Dehai Wu,
| |
Collapse
|
7
|
Lan T, Qian S, Tang C, Gao J. Role of Immune Cells in Biliary Repair. Front Immunol 2022; 13:866040. [PMID: 35432349 PMCID: PMC9005827 DOI: 10.3389/fimmu.2022.866040] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/08/2022] [Indexed: 02/06/2023] Open
Abstract
The biliary system is comprised of cholangiocytes and plays an important role in maintaining liver function. Under normal conditions, cholangiocytes remain in the stationary phase and maintain a very low turnover rate. However, the robust biliary repair is initiated in disease conditions, and different repair mechanisms can be activated depending on the pathological changes. During biliary disease, immune cells including monocytes, lymphocytes, neutrophils, and mast cells are recruited to the liver. The cellular interactions between cholangiocytes and these recruited immune cells as well as hepatic resident immune cells, including Kupffer cells, determine disease outcomes. However, the role of immune cells in the initiation, regulation, and suspension of biliary repair remains elusive. The cellular processes of cholangiocyte proliferation, progenitor cell differentiation, and hepatocyte-cholangiocyte transdifferentiation during biliary diseases are reviewed to manifest the underlying mechanism of biliary repair. Furthermore, the potential role of immune cells in crucial biliary repair mechanisms is highlighted. The mechanisms of biliary repair in immune-mediated cholangiopathies, inherited cholangiopathies, obstructive cholangiopathies, and cholangiocarcinoma are also summarized. Additionally, novel techniques that could clarify the underlying mechanisms of biliary repair are displayed. Collectively, this review aims to deepen the understanding of the mechanisms of biliary repair and contributes potential novel therapeutic methods for treating biliary diseases.
Collapse
Affiliation(s)
- Tian Lan
- Lab 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
| | - Shuaijie Qian
- Lab 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
| | - Chengwei Tang
- Lab 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
| | - Jinhang Gao
- Lab 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
| |
Collapse
|
8
|
Pham L, Kennedy L, Baiocchi L, Meadows V, Ekser B, Kundu D, Zhou T, Sato K, Glaser S, Ceci L, Alpini G, Francis H. Mast cells in liver disease progression: An update on current studies and implications. Hepatology 2022; 75:213-218. [PMID: 34435373 PMCID: PMC9276201 DOI: 10.1002/hep.32121] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/24/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Linh Pham
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA,Department of Science and Mathematics, Texas A&M University–Central Texas, Killeen, Texas, USA
| | - Lindsey Kennedy
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA,Richard L. Roudebush VA Medical Center, Indianapolis, Indiana, USA
| | | | - Vik Meadows
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Burcin Ekser
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Debjyoti Kundu
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Tianhao Zhou
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Keisaku Sato
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Shannon Glaser
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Ludovica Ceci
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Gianfranco Alpini
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA,Richard L. Roudebush VA Medical Center, Indianapolis, Indiana, USA
| | - Heather Francis
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA,Richard L. Roudebush VA Medical Center, Indianapolis, Indiana, USA
| |
Collapse
|
9
|
Ahmad A, Ansari MM, AlAsmari AF, Ali N, Maqbool MT, Raza SS, Khan R. Dose dependent safety implications and acute intravenous toxicity of aminocellulose-grafted-polycaprolactone coated gelatin nanoparticles in mice. Int J Biol Macromol 2021; 192:1150-1159. [PMID: 34653441 DOI: 10.1016/j.ijbiomac.2021.10.028] [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: 05/29/2021] [Revised: 09/27/2021] [Accepted: 10/03/2021] [Indexed: 10/20/2022]
Abstract
Polymeric nanoparticles (NPs) are the most widely researched nanoformulations and gained broad acceptance in nanotherapeutics for targeted drug delivery and theranostics. However, lack of regulations, guidelines, harmonized standards, and limitations with their employability in clinical circumstances necessitates an in-depth understanding of their toxicology. Here, we examined the in-vivo toxicity of core-shell polymeric NPs made up of gelatin core coated with an outer layer of aminocellulose-grafted polycaprolactone (PCL-AC) synthesized for drug delivery purposes in inflammatory disorders. Nanoparticles were administered intravenously in Swiss albino mice, in multiple dosing (10, 25, and 50 mg/kg body weight) and outcomes of serum biochemistry analysis and histopathology evaluation exhibited that the highest 50 mg/kg administration of NPs altered biochemistry and histopathology aspects of vital organs, while doses of 10 and 25 mg/kg were safe and biocompatible. Further, mast cell (toluidine blue) staining confirmed that administration of the highest dose enhanced mast cell infiltration in tissues of vital organs, while lower doses did not exhibit any of these alterations. Therefore, the results of the present study establish that the NPs disposal in-vivo culminates into alterations in organ structure and function consequences such that lower doses are quite biocompatible and do not demonstrate any structural or functional toxicity while some toxicological effects start appearing at the highest dose.
Collapse
Affiliation(s)
- Anas Ahmad
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Sector-81, Knowledge City, Sahibzada Ajit Singh Nagar, Punjab Pin 140306, India
| | - Md Meraj Ansari
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Sector-81, Knowledge City, Sahibzada Ajit Singh Nagar, Punjab Pin 140306, India
| | - Abdullah F AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P. O. Box 55760, Riyadh 11451, Saudi Arabia
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P. O. Box 55760, Riyadh 11451, Saudi Arabia
| | - Mir Tahir Maqbool
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, MS 38677, USA
| | - Syed Shadab Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Sarfarazganj, Lucknow 226003, Uttar Pradesh, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Sector-81, Knowledge City, Sahibzada Ajit Singh Nagar, Punjab Pin 140306, India.
| |
Collapse
|
10
|
Wang YJ, Downey MA, Choi S, Shoup TM, Elmaleh DR. Cromolyn platform suppresses fibrosis and inflammation, promotes microglial phagocytosis and neurite outgrowth. Sci Rep 2021; 11:22161. [PMID: 34772945 PMCID: PMC8589953 DOI: 10.1038/s41598-021-00465-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/07/2021] [Indexed: 12/18/2022] Open
Abstract
Neurodegenerative diseases are characterized by chronic neuroinflammation and may perpetuate ongoing fibrotic reactions within the central nervous system. Unfortunately, there is no therapeutic available that treats neurodegenerative inflammation and its sequelae. Here we utilize cromolyn, a mast cell inhibitor with anti-inflammatory capabilities, and its fluorinated analogue F-cromolyn to study fibrosis-related protein regulation and secretion downstream of neuroinflammation and their ability to promote microglial phagocytosis and neurite outgrowth. In this report, RNA-seq analysis shows that administration of the pro-inflammatory cytokine TNF-α to HMC3 human microglia results in a robust upregulation of fibrosis-associated genes. Subsequent treatment with cromolyn and F-cromolyn resulted in reduced secretion of collagen XVIII, fibronectin, and tenascin-c. Additionally, we show that cromolyn and F-cromolyn reduce pro-inflammatory proteins PLP1, PELP1, HSP90, IL-2, GRO-α, Eotaxin, and VEGF-Α, while promoting secretion of anti-inflammatory IL-4 in HMC3 microglia. Furthermore, cromolyn and F-cromolyn augment neurite outgrowth in PC12 neuronal cells in concert with nerve growth factor. Treatment also differentially altered secretion of neurogenesis-related proteins TTL, PROX1, Rab35, and CSDE1 in HMC3 microglia. Finally, iPSC-derived human microglia more readily phagocytose Aβ42 with cromolyn and F-cromolyn relative to controls. We propose the cromolyn platform targets multiple proteins upstream of PI3K/Akt/mTOR, NF-κB, and GSK-3β signaling pathways to affect cytokine, chemokine, and fibrosis-related protein expression.
Collapse
Affiliation(s)
| | | | - Sungwoon Choi
- Department of New Drug Discovery, Chungnam National University, Daejeon, South Korea
| | - Timothy M Shoup
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129-2060, USA
| | - David R Elmaleh
- AZTherapies, Inc., Boston, MA, USA.
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129-2060, USA.
| |
Collapse
|
11
|
Kennedy L, Meadows V, Sybenga A, Demieville J, Chen L, Hargrove L, Ekser B, Dar W, Ceci L, Kundu D, Kyritsi K, Pham L, Zhou T, Glaser S, Meng F, Alpini G, Francis H. Mast Cells Promote Nonalcoholic Fatty Liver Disease Phenotypes and Microvesicular Steatosis in Mice Fed a Western Diet. Hepatology 2021; 74:164-182. [PMID: 33434322 PMCID: PMC9271361 DOI: 10.1002/hep.31713] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND AND AIMS Nonalcoholic fatty liver disease (NAFLD) is simple steatosis but can develop into nonalcoholic steatohepatitis (NASH), characterized by liver inflammation, fibrosis, and microvesicular steatosis. Mast cells (MCs) infiltrate the liver during cholestasis and promote ductular reaction (DR), biliary senescence, and liver fibrosis. We aimed to determine the effects of MC depletion during NAFLD/NASH. APPROACH AND RESULTS Wild-type (WT) and KitW-sh (MC-deficient) mice were fed a control diet (CD) or a Western diet (WD) for 16 weeks; select WT and KitW-sh WD mice received tail vein injections of MCs 2 times per week for 2 weeks prior to sacrifice. Human samples were collected from normal, NAFLD, or NASH mice. Cholangiocytes from WT WD mice and human NASH have increased insulin-like growth factor 1 expression that promotes MC migration/activation. Enhanced MC presence was noted in WT WD mice and human NASH, along with increased DR. WT WD mice had significantly increased steatosis, DR/biliary senescence, inflammation, liver fibrosis, and angiogenesis compared to WT CD mice, which was significantly reduced in KitW-sh WD mice. Loss of MCs prominently reduced microvesicular steatosis in zone 1 hepatocytes. MC injection promoted WD-induced biliary and liver damage and specifically up-regulated microvesicular steatosis in zone 1 hepatocytes. Aldehyde dehydrogenase 1 family, member A3 (ALDH1A3) expression is reduced in WT WD mice and human NASH but increased in KitW-sh WD mice. MicroRNA 144-3 prime (miR-144-3p) expression was increased in WT WD mice and human NASH but reduced in KitW-sh WD mice and was found to target ALDH1A3. CONCLUSIONS MCs promote WD-induced biliary and liver damage and may promote microvesicular steatosis development during NAFLD progression to NASH through miR-144-3p/ALDH1A3 signaling. Inhibition of MC activation may be a therapeutic option for NAFLD/NASH treatment.
Collapse
Affiliation(s)
- Lindsey Kennedy
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Vik Meadows
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Amelia Sybenga
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Jennifer Demieville
- Central Texas Veterans Health Care System, Texas A&M University College of Medicine, Bryan, TX
| | - Lixian Chen
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Laura Hargrove
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX
| | - Burcin Ekser
- Department of Transplant Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - Wasim Dar
- Division of Immunology and Organ Transplantation, Department of Surgery, University of Texas Health Science Center at Houston, Houston, TX
| | - Ludovica Ceci
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Debjyoti Kundu
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Konstantina Kyritsi
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Linh Pham
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Tianhao Zhou
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX
| | - Shannon Glaser
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX
| | - Fanyin Meng
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN,Richard L. Roudebush VA Medical Center, Indiana University School of Medicine, Indianapolis, IN
| | - Gianfranco Alpini
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN,Richard L. Roudebush VA Medical Center, Indiana University School of Medicine, Indianapolis, IN
| | - Heather Francis
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN,Richard L. Roudebush VA Medical Center, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
12
|
Kyritsi K, Kennedy L, Meadows V, Hargrove L, Demieville J, Pham L, Sybenga A, Kundu D, Cerritos K, Meng F, Alpini G, Francis H. Mast Cells Induce Ductular Reaction Mimicking Liver Injury in Mice Through Mast Cell-Derived Transforming Growth Factor Beta 1 Signaling. Hepatology 2021; 73:2397-2410. [PMID: 32761972 PMCID: PMC7864988 DOI: 10.1002/hep.31497] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/15/2020] [Accepted: 06/28/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Following liver injury, mast cells (MCs) migrate into the liver and are activated in patients with cholestasis. Inhibition of MC mediators decreases ductular reaction (DR) and liver fibrosis. Transforming growth factor beta 1 (TGF-β1) contributes to fibrosis and promotes liver disease. Our aim was to demonstrate that reintroduction of MCs induces cholestatic injury through TGF-β1. APPROACH AND RESULTS Wild-type, KitW-sh (MC-deficient), and multidrug resistance transporter 2/ABC transporter B family member 2 knockout mice lacking l-histidine decarboxylase were injected with vehicle or PKH26-tagged murine MCs pretreated with 0.01% dimethyl sulfoxide (DMSO) or the TGF-β1 receptor inhibitor (TGF-βRi), LY2109761 (10 μM) 3 days before sacrifice. Hepatic damage was assessed by hematoxylin and eosin (H&E) and serum chemistry. Injected MCs were detected in liver, spleen, and lung by immunofluorescence (IF). DR was measured by cytokeratin 19 (CK-19) immunohistochemistry and F4/80 staining coupled with real-time quantitative PCR (qPCR) for interleukin (IL)-1β, IL-33, and F4/80; biliary senescence was evaluated by IF or qPCR for p16, p18, and p21. Fibrosis was evaluated by sirius red/fast green staining and IF for synaptophysin 9 (SYP-9), desmin, and alpha smooth muscle actin (α-SMA). TGF-β1 secretion/expression was measured by enzyme immunoassay and qPCR. Angiogenesis was detected by IF for von Willebrand factor and vascular endothelial growth factor C qPCR. In vitro, MC-TGF-β1 expression/secretion were measured after TGF-βRi treatment; conditioned medium was collected. Cholangiocytes and hepatic stellate cells (HSCs) were treated with MC-conditioned medium, and biliary proliferation/senescence was measured by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium and qPCR; HSC activation evaluated for α-SMA, SYP-9, and collagen type-1a expression. MC injection recapitulates cholestatic liver injury characterized by increased DR, fibrosis/TGF-β1 secretion, and angiogenesis. Injection of MC-TGF-βRi reversed these parameters. In vitro, MCs induce biliary proliferation/senescence and HSC activation that was reversed with MCs lacking TGF-β1. CONCLUSIONS Our study demonstrates that reintroduction of MCs mimics cholestatic liver injury and that MC-derived TGF-β1 may be a target in chronic cholestatic liver disease.
Collapse
Affiliation(s)
- Konstantina Kyritsi
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine Research
| | - Lindsey Kennedy
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine Research
| | - Vik Meadows
- Richard L. Roudebush VA Medical Center, Indiana University School of Medicine Research,Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine Research
| | - Laura Hargrove
- Texas A&M University Health Science Center, Texas A&M University-Central Texas
| | | | - Linh Pham
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine Research
| | | | - Debjyoti Kundu
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine Research
| | - Karla Cerritos
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine Research
| | - Fanyin Meng
- Richard L. Roudebush VA Medical Center, Indiana University School of Medicine Research,Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine Research
| | - Gianfranco Alpini
- Richard L. Roudebush VA Medical Center, Indiana University School of Medicine Research,Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine Research
| | - Heather Francis
- Richard L. Roudebush VA Medical Center, Indiana University School of Medicine Research,Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine Research
| |
Collapse
|
13
|
Park Y, Ahn JH, Lee TK, Kim B, Tae HJ, Park JH, Shin MC, Cho JH, Won MH. Therapeutic hypothermia reduces inflammation and oxidative stress in the liver after asphyxial cardiac arrest in rats. Acute Crit Care 2020; 35:286-295. [PMID: 33423440 PMCID: PMC7808856 DOI: 10.4266/acc.2020.00304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/12/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Few studies have evaluated the effects of hypothermia on cardiac arrest (CA)-induced liver damage. This study aimed to investigate the effects of hypothermic therapy on the liver in a rat model of asphyxial cardiac arrest (ACA). METHODS Rats were subjected to 5-minute ACA followed by return of spontaneous circulation (RoSC). Body temperature was controlled at 33°C±0.5°C or 37°C±0.5°C for 4 hours after RoSC in the hypothermia group and normothermia group, respectively. Liver tissues in each group were collected at 6 hours, 12 hours, 1 day, and 2 days after RoSC. To examine hepatic inflammation, mast cells were stained with toluidine blue. Superoxide anion radical production was evaluated using dihydroethidium fluorescence straining and expression of endogenous antioxidants (superoxide dismutase 1 [SOD1] and SOD2) was examined using immunohistochemistry. RESULTS There were significantly more mast cells in the livers of the normothermia group with ACA than in the hypothermia group with ACA. Gradual increase in superoxide anion radical production was found with time in the normothermia group with ACA, but production was significantly suppressed in the hypothermia group with ACA relative to the normothermia group with ACA. SOD1 and SOD2 levels were higher in the hypothermia group with ACA than in the normothermia group with ACA. CONCLUSIONS Experimental hypothermic treatment after ACA significantly inhibited inflammation and superoxide anion radical production in the rat liver, indicating that this treatment enhanced or maintained expression of antioxidants. Our findings suggest that hypothermic therapy after CA can reduce mast cell-mediated inflammation through regulation of oxidative stress and the expression of antioxidants in the liver.
Collapse
Affiliation(s)
- Yoonsoo Park
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Ji Hyeon Ahn
- Department of Physical Therapy, College of Health Science, Youngsan University, Yangsan, Korea.,Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Tae-Kyeong Lee
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Korea
| | - Bora Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Hyun-Jin Tae
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Korea
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju, Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Korea
| |
Collapse
|
14
|
Kundu D, Kennedy L, Meadows V, Baiocchi L, Alpini G, Francis H. The Dynamic Interplay Between Mast Cells, Aging/Cellular Senescence, and Liver Disease. Gene Expr 2020; 20:77-88. [PMID: 32727636 PMCID: PMC7650013 DOI: 10.3727/105221620x15960509906371] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mast cells are key players in acute immune responses that are evidenced by degranulation leading to a heightened allergic response. Activation of mast cells can trigger a number of different pathways contributing to metabolic conditions and disease progression. Aging results in irreversible physiological changes affecting all organs, including the liver. The liver undergoes senescence, changes in protein expression, and cell signaling phenotypes during aging, which regulate disease progression. Cellular senescence contributes to the age-related changes. Unsurprisingly, mast cells also undergo age-related changes in number, localization, and activation throughout their lifetime, which adversely affects the etiology and progression of many physiological conditions including liver diseases. In this review, we discuss the role of mast cells during aging, including features of aging (e.g., senescence) in the context of biliary diseases such as primary biliary cholangitis and primary sclerosing cholangitis and nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Debjyoti Kundu
- *Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lindsey Kennedy
- *Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Vik Meadows
- *Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Leonardo Baiocchi
- †Department of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Gianfranco Alpini
- *Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- ‡Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Heather Francis
- *Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- ‡Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
15
|
Ahmad A, Ansari MM, Kumar A, Vyawahare A, Mishra RK, Jayamurugan G, Raza SS, Khan R. Comparative acute intravenous toxicity study of triple polymer-layered magnetic nanoparticles with bare magnetic nanoparticles in Swiss albino mice. Nanotoxicology 2020; 14:1362-1380. [DOI: 10.1080/17435390.2020.1829144] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Anas Ahmad
- Institute of Nano Science and Technology, Habitat Centre, Mohali, India
| | - Md. Meraj Ansari
- Institute of Nano Science and Technology, Habitat Centre, Mohali, India
| | - Ajay Kumar
- Institute of Nano Science and Technology, Habitat Centre, Mohali, India
| | - Akshay Vyawahare
- Institute of Nano Science and Technology, Habitat Centre, Mohali, India
| | | | | | - Syed Shadab Raza
- Laboratory for Stem Cell and Restorative Neurology, Department of Biotechnology, Era’s Lucknow Medical College Hospital, Lucknow, India
- Department of Stem Cell Biology and Regenerative Medicine, Era University, Lucknow, India
| | - Rehan Khan
- Institute of Nano Science and Technology, Habitat Centre, Mohali, India
| |
Collapse
|
16
|
Weiskirchen R, Meurer SK, Liedtke C, Huber M. Mast Cells in Liver Fibrogenesis. Cells 2019; 8:E1429. [PMID: 31766207 PMCID: PMC6912398 DOI: 10.3390/cells8111429] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/05/2019] [Accepted: 11/10/2019] [Indexed: 01/10/2023] Open
Abstract
Mast cells (MCs) are immune cells of the myeloid lineage that are present in the connective tissue throughout the body and in mucosa tissue. They originate from hematopoietic stem cells in the bone marrow and circulate as MC progenitors in the blood. After migration to various tissues, they differentiate into their mature form, which is characterized by a phenotype containing large granules enriched in a variety of bioactive compounds, including histamine and heparin. These cells can be activated in a receptor-dependent and -independent manner. Particularly, the activation of the high-affinity immunoglobulin E (IgE) receptor, also known as FcεRI, that is expressed on the surface of MCs provoke specific signaling cascades that leads to intracellular calcium influx, activation of different transcription factors, degranulation, and cytokine production. Therefore, MCs modulate many aspects in physiological and pathological conditions, including wound healing, defense against pathogens, immune tolerance, allergy, anaphylaxis, autoimmune defects, inflammation, and infectious and other disorders. In the liver, MCs are mainly associated with connective tissue located in the surrounding of the hepatic arteries, veins, and bile ducts. Recent work has demonstrated a significant increase in MC number during hepatic injury, suggesting an important role of these cells in liver disease and progression. In the present review, we summarize aspects of MC function and mediators in experimental liver injury, their interaction with other hepatic cell types, and their contribution to the pathogenesis of fibrosis.
Collapse
Affiliation(s)
- Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), University Hospital, RWTH Aachen University, D-52074 Aachen, Germany;
| | - Steffen K. Meurer
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), University Hospital, RWTH Aachen University, D-52074 Aachen, Germany;
| | - Christian Liedtke
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany;
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, D-52074 Aachen, Germany
| |
Collapse
|
17
|
Tolefree JA, Garcia AJ, Farrell J, Meadows V, Kennedy L, Hargrove L, Demieville J, Francis N, Mirabel J, Francis H. Alcoholic liver disease and mast cells: What's your gut got to do with it? LIVER RESEARCH 2019. [DOI: 10.1016/j.livres.2019.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
18
|
Tanino T, Bando T, Nojiri Y, Okada Y, Nagai N, Ueda Y, Sakurai E. Hepatic cytochrome P450 metabolism suppressed by mast cells in type 1 allergic mice. Biochem Pharmacol 2018; 158:318-326. [PMID: 30395837 DOI: 10.1016/j.bcp.2018.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/01/2018] [Indexed: 12/11/2022]
Abstract
Mast cells and Kupffer cells secrete interleukin (IL)-1β, interferon (IFN)-γ, and tumor necrosis factor (TNF)-α, which stimulate excess nitric oxide (NO) producing-inducible NO synthase (iNOS). Unlike Kupffer cells, immunoglobulin E-sensitized mast cells elicit sustained NO production. We investigated the participation of mast cell-released NO and cytokine-derived iNOS activation in type 1 allergy-suppressed hepatic cytochrome P450 (CYP) metabolism. Aminoguanidine, a selective iNOS inhibitor, completely suppressed serum nitrate plus nitrite (NOx) concentrations after primary and secondary sensitization of ICR mice and markedly attenuated allergy-suppressed hepatic CYP1A2, CYP2C, CYP2E1, and CYP3A activities. In the liver, primary and secondary sensitization enhanced iNOS-stimulating IFN-γ (5-15-fold) and TNF-α (3-5-fold) mRNA levels more than IL-1β (2-fold) and F4/80-positive Kupffer cell (2-fold) mRNA levels. When mast cell-deficient (-/-) mice were sensitized, hepatic CYP activities were not suppressed. Serum NOx levels in the sensitized -/- mice were similar with those in saline-treated ICR and -/- mice. In the liver of -/- mice, secondary sensitization markedly enhanced mRNA expression of iNOS (20-fold), IFN-γ (15-fold), and TNF-α (3-fold). However, hepatic total NOS activities in -/- mice were not significantly different between saline treatment and sensitization. Similarly, primary and secondary ICR mice did not significantly enhance total NOS activities in the liver and hepatocytes. The total NOS activities observed did not relate to the high levels of iNOS, IFN-γ, and TNF-α mRNA in the liver. Hepatic c-kit-positive mast cells in sensitized ICR mice were maintained at control levels. Therefore, our data suggest that mast cell-released NO participates in type 1 allergy-suppressed CYP1A2, CYP2C, CYP2E1, and CYP3A metabolism.
Collapse
Affiliation(s)
- Tadatoshi Tanino
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Bouji Nishihama, Yamashiro-cho, Tokushima, Tokushima 770-8514, Japan
| | - Toru Bando
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Bouji Nishihama, Yamashiro-cho, Tokushima, Tokushima 770-8514, Japan
| | - Yukie Nojiri
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Bouji Nishihama, Yamashiro-cho, Tokushima, Tokushima 770-8514, Japan
| | - Yuna Okada
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Bouji Nishihama, Yamashiro-cho, Tokushima, Tokushima 770-8514, Japan
| | - Noriaki Nagai
- Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, Osaka 577-8502, Japan
| | - Yukari Ueda
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Bouji Nishihama, Yamashiro-cho, Tokushima, Tokushima 770-8514, Japan
| | - Eiichi Sakurai
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Bouji Nishihama, Yamashiro-cho, Tokushima, Tokushima 770-8514, Japan.
| |
Collapse
|
19
|
Jiang L, Fang P, Septer S, Apte U, Pritchard MT. Inhibition of Mast Cell Degranulation With Cromolyn Sodium Exhibits Organ-Specific Effects in Polycystic Kidney (PCK) Rats. Int J Toxicol 2018; 37:308-326. [PMID: 29862868 PMCID: PMC6027616 DOI: 10.1177/1091581818777754] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Autosomal recessive polycystic kidney disease (ARPKD) is a monogenic disease characterized by development of hepatorenal cysts, pericystic fibrosis, and inflammation. Previous studies show that mast cell (MC) mediators such as histamine induce proliferation of cholangiocytes. We observed robust MC accumulation around liver cysts, but not kidney cysts, in polycystic kidney (PCK) rats (an animal model of ARPKD). Therefore, we hypothesized that MCs contribute to hepatic cyst growth in ARPKD. To test this hypothesis, we treated PCK rats with 1 of 2 different MC stabilizers, cromolyn sodium (CS) or ketotifen, or saline. The CS treatment decreased MC degranulation in the liver and reduced serum tryptase (an MC granule component). Interestingly, we observed an increase in liver to body weight ratio after CS treatment paralleled by a significant increase in individual cyst size. Hepatic fibrosis was not affected by CS treatment. The CS treatment increased hepatic cyst wall epithelial cell (CWEC) proliferation and decreased cell death. Ketotifen treatment also increased hepatic cyst size. In vitro, CS treatment did not affect proliferation of isolated hepatic CWECs from PCK rats. In contrast, CS decreased kidney to body weight ratio paralleled by a significant decrease in individual cyst size. The percentage of kidney to body weight ratio was strongly correlated with serum renin (an MC granule component). Ketotifen did not affect kidney cyst growth. Collectively, these data suggest that CS affects hepatic and renal cyst growth differently in PCK rats. Moreover, CS may be beneficial to renal cystic disease but may exacerbate hepatic cyst growth in ARPKD.
Collapse
Affiliation(s)
- Lu Jiang
- 1 Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Pingping Fang
- 1 Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Seth Septer
- 2 Department of Pediatric Gastroenterology, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, CO, USA
| | - Udayan Apte
- 1 Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
- 3 Liver Center, University of Kansas Medical Center, Kansas City, KS, USA
- 4 The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Michele T Pritchard
- 1 Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
- 3 Liver Center, University of Kansas Medical Center, Kansas City, KS, USA
- 4 The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
20
|
Jarido V, Kennedy L, Hargrove L, Demieville J, Thomson J, Stephenson K, Francis H. The emerging role of mast cells in liver disease. Am J Physiol Gastrointest Liver Physiol 2017; 313:G89-G101. [PMID: 28473331 PMCID: PMC5582878 DOI: 10.1152/ajpgi.00333.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 04/12/2017] [Accepted: 04/24/2017] [Indexed: 01/31/2023]
Abstract
The depth of our knowledge regarding mast cells has widened exponentially in the last 20 years. Once thought to be only important for allergy-mediated events, mast cells are now recognized to be important regulators of a number of pathological processes. The revelation that mast cells can influence organs, tissues, and cells has increased interest in mast cell research during liver disease. The purpose of this review is to refresh the reader's knowledge of the development, type, and location of mast cells and to review recent work that demonstrates the role of hepatic mast cells during diseased states. This review focuses primarily on liver diseases and mast cells during autoimmune disease, hepatitis, fatty liver disease, liver cancer, and aging in the liver. Overall, these studies demonstrate the potential role of mast cells in disease progression.
Collapse
Affiliation(s)
- Veronica Jarido
- Baylor Scott & White Health and Medicine, Temple, Texas; and
| | - Lindsey Kennedy
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Texas A & M Health Science Center, Temple, Texas
| | | | | | - Joanne Thomson
- Research, Central Texas Veterans Health Care System, Temple, Texas
| | | | - Heather Francis
- Research, Central Texas Veterans Health Care System, Temple, Texas;
- Baylor Scott & White Health and Medicine, Temple, Texas; and
- Texas A & M Health Science Center, Temple, Texas
| |
Collapse
|
21
|
Thomson J, Hargrove L, Kennedy L, Demieville J, Francis H. Cellular crosstalk during cholestatic liver injury. LIVER RESEARCH 2017; 1:26-33. [PMID: 29552372 PMCID: PMC5854144 DOI: 10.1016/j.livres.2017.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The functions of the liver are very diverse. From detoxifying blood to storing glucose in the form of glycogen and producing bile to facilitate fat digestion, the liver is a very active and important organ. The liver is comprised of many varied cell types whose functions are equally diverse. Cholangiocytes line the biliary tree and aid in transporting and adjusting the composition of bile as it travels to the gallbladder. Hepatic stellate cells and portal fibroblasts are located in different areas within the liver architecture, but both contribute to the development of fibrosis upon activation after liver injury. Vascular cells, including those that constitute the peribiliary vascular plexus, are involved in functions other than blood delivery to and from the liver, such as supporting the growth of the biliary tree during development. Mast cells are normally found in healthy livers but in very low numbers. However, after injury, mast cell numbers greatly increase as they infiltrate and release factors that exacerbate the fibrotic response. While not an all-inclusive list, these cells have individual roles within the liver, but they are also able to communicate with each other by cellular crosstalk. In this review, we examine some of these pathways that can lead to an increase in the homeostatic dysfunction seen in liver injury.
Collapse
Affiliation(s)
- Joanne Thomson
- Research, Central Texas Veterans Healthcare System, TX, USA
| | - Laura Hargrove
- Medicine, Texas A&M Health Science Center, Temple, TX, USA
| | - Lindsey Kennedy
- Research, Central Texas Veterans Healthcare System, TX, USA
- Medicine, Texas A&M Health Science Center, Temple, TX, USA
| | | | - Heather Francis
- Research, Central Texas Veterans Healthcare System, TX, USA
- Digestive Disease Research Center, Baylor Scott & White Health, TX, USA
- Medicine, Texas A&M Health Science Center, Temple, TX, USA
| |
Collapse
|