1
|
Matsuda T, Kaji K, Nishimura N, Asada S, Koizumi A, Tanaka M, Yorioka N, Tsuji Y, Kitagawa K, Sato S, Namisaki T, Akahane T, Yoshiji H. Cabozantinib prevents the progression of metabolic dysfunction-associated steatohepatitis by inhibiting the activation of hepatic stellate cell and macrophage and attenuating angiogenic activity. Heliyon 2024; 10:e38647. [PMID: 39398008 PMCID: PMC11470516 DOI: 10.1016/j.heliyon.2024.e38647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 09/05/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024] Open
Abstract
Cabozantinib, a multiple tyrosine kinase inhibitor targeting AXL, vascular endothelial growth factor receptor (VEGFR), and MET, is used clinically to treat certain cancers, including hepatocellular carcinoma. This study aimed to assess the impact of cabozantinib on liver fibrosis and hepatocarcinogenesis in a rat model of metabolic dysfunction-associated steatohepatitis (MASH). MASH-based liver fibrosis and hepatocarcinogenesis were induced in rats by feeding them a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) for eight and 16 weeks, respectively. Cabozantinib (1 or 2 mg/kg, daily) was administered concurrently with the diet in the fibrosis model and after eight weeks in the carcinogenesis model. Treatment with cabozantinib significantly attenuated hepatic inflammation and fibrosis without affecting hepatocyte steatosis and ballooning in CDAHFD-fed rats. Cabozantinib-treated rats exhibited a marked reduction in α-smooth muscle actin+ activated hepatic stellate cell (HSC) expansion, CD68+ macrophage infiltration, and CD34+ pathological angiogenesis, along with reduced hepatic AXL, VEGF, and VEGFR2 expression. Consistently, cabozantinib downregulated the hepatic expression of profibrogenic markers (Acta2, Col1a1, Tgfb1), inflammatory cytokines (Tnfa, Il1b, Il6), and proangiogenic markers (Vegfa, Vwf, Ang2). In a cell-based assay of human activated HSCs, cabozantinib inhibited Akt activation induced by GAS6, a ligand of AXL, leading to reduced cell proliferation and profibrogenic activity. Cabozantinib also suppressed lipopolysaccharide-induced proinflammatory responses in human macrophages, VEGFA-induced collagen expression and proliferation in activated HSCs, and VEGFA-stimulated proliferation in vascular endothelial cells. Meanwhile, administration of cabozantinib did not affect Ki67+ hepatocyte proliferation or serum albumin levels, indicating no negative impact on regenerative capacity. Treatment with cabozantinib also reduced the placental glutathione transferase+ preneoplastic lesions in CDAHFD-fed rats. In conclusion, cabozantinib shows promise as a novel option for preventing MASH progression.
Collapse
Affiliation(s)
- Takuya Matsuda
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kosuke Kaji
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Norihisa Nishimura
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Shohei Asada
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Aritoshi Koizumi
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Misako Tanaka
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Nobuyuki Yorioka
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yuki Tsuji
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Koh Kitagawa
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Shinya Sato
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Tadashi Namisaki
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Takemi Akahane
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Hitoshi Yoshiji
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| |
Collapse
|
2
|
Tutusaus A, Morales A, García de Frutos P, Marí M. GAS6/TAM Axis as Therapeutic Target in Liver Diseases. Semin Liver Dis 2024; 44:99-114. [PMID: 38395061 PMCID: PMC11027478 DOI: 10.1055/a-2275-0408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
TAM (TYRO3, AXL, and MERTK) protein tyrosine kinase membrane receptors and their vitamin K-dependent ligands GAS6 and protein S (PROS) are well-known players in tumor biology and autoimmune diseases. In contrast, TAM regulation of fibrogenesis and the inflammation mechanisms underlying metabolic dysfunction-associated steatohepatitis (MASH), cirrhosis, and, ultimately, liver cancer has recently been revealed. GAS6 and PROS binding to phosphatidylserine exposed in outer membranes of apoptotic cells links TAMs, particularly MERTK, with hepatocellular damage. In addition, AXL and MERTK regulate the development of liver fibrosis and inflammation in chronic liver diseases. Acute hepatic injury is also mediated by the TAM system, as recent data regarding acetaminophen toxicity and acute-on-chronic liver failure have uncovered. Soluble TAM-related proteins, mainly released from activated macrophages and hepatic stellate cells after hepatic deterioration, are proposed as early serum markers for disease progression. In conclusion, the TAM system is becoming an interesting pharmacological target in liver pathology and a focus of future biomedical research in this field.
Collapse
Affiliation(s)
- Anna Tutusaus
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, Barcelona, Spain
| | - Albert Morales
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, Barcelona, Spain
| | - Pablo García de Frutos
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Cardiovasculares (CIBERCV), Barcelona, Comunidad de Madrid, Spain
| | - Montserrat Marí
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, Barcelona, Spain
| |
Collapse
|
3
|
Apostolo D, Ferreira LL, Vincenzi F, Vercellino N, Minisini R, Latini F, Ferrari B, Burlone ME, Pirisi M, Bellan M. From MASH to HCC: the role of Gas6/TAM receptors. Front Immunol 2024; 15:1332818. [PMID: 38298195 PMCID: PMC10827955 DOI: 10.3389/fimmu.2024.1332818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is the replacement term for what used to be called nonalcoholic steatohepatitis (NASH). It is characterized by inflammation and injury of the liver in the presence of cardiometabolic risk factors and may eventually result in the development of hepatocellular carcinoma (HCC), the most common form of primary liver cancer. Several pathogenic mechanisms are involved in the transition from MASH to HCC, encompassing metabolic injury, inflammation, immune dysregulation and fibrosis. In this context, Gas6 (Growth Arrest-Specific 6) and TAM (Tyro3, Axl, and MerTK) receptors may play important roles. The Gas6/TAM family is involved in the modulation of inflammation, lipid metabolism, fibrosis, tumor progression and metastasis, processes which play an important role in the pathophysiology of acute and chronic liver diseases. In this review, we discuss MASH-associated HCC and the potential involvement of the Gas6/TAM system in disease development and progression. In addition, since therapeutic strategies for MASH and HCC are limited, we also speculate regarding possible future treatments involving the targeting of Gas6 or TAM receptors.
Collapse
Affiliation(s)
- Daria Apostolo
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Luciana L. Ferreira
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Federica Vincenzi
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Nicole Vercellino
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Rosalba Minisini
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Federico Latini
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Barbara Ferrari
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Michela E. Burlone
- Department of Internal Medicine, Azienda Ospedaliero-Universitaria Maggiore Della Carità, Novara, Italy
| | - Mario Pirisi
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- Department of Internal Medicine, Azienda Ospedaliero-Universitaria Maggiore Della Carità, Novara, Italy
- Center on Autoimmune and Allergic Diseases, Università del Piemonte Orientale, Novara, Italy
| | - Mattia Bellan
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- Department of Internal Medicine, Azienda Ospedaliero-Universitaria Maggiore Della Carità, Novara, Italy
- Center on Autoimmune and Allergic Diseases, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
4
|
Li X, Wang Q, Ai L, Cheng K. Unraveling the activation process and core driver genes of HSCs during cirrhosis by single-cell transcriptome. Exp Biol Med (Maywood) 2023; 248:1414-1424. [PMID: 37674431 PMCID: PMC10657590 DOI: 10.1177/15353702231191109] [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: 12/23/2022] [Accepted: 03/11/2023] [Indexed: 09/08/2023] Open
Abstract
Worldwide, cirrhosis is a common cause of death, manifesting itself as fibrosis of the liver tissue. When the liver is damaged, the liver produces fibrotic, proliferative myofibroblasts, which are formed by the differentiation of activated hepatic stellate cells. There are no effective antifibrotic treatment options. To deeply explore the activation process of hepatic stellate cells (HSCs) and to discover better therapeutic target genes, single-cell RNA sequencing data on 13 non-cirrhotic liver tissue samples and 10 cirrhotic liver tissue samples were analyzed. We identified activated HSCs from the mesenchymal cell population with high expression of ACTA2. By pseudo-time analysis, we found that the key genes for the differentiation of HSCs into myofibroblasts were C3, CCDC80, COL1A1, COL3A1, DCN, FBLN1, IGFBP3, MXRA5, SERPINE1, and MYH11. Then, we found that the main regulators of HSCs from inactive to activated state were NTF3, NTRK3, NTRK2, JAG1, NOTCH3, ESAM, and CD46 by cell-cell communication analysis. In addition, we found that the top2 hub genes of activated HSCs were CRIP1 and ACTA2. The experimental results show that the top2 hub genes were significantly overexpressed in cirrhotic samples. Our work dissected key intercellular regulators and core driver genes during hepatic stellate cell activation during cirrhosis through single-cell transcriptome data analysis, providing a research strategy to discover rational therapeutic targets for cirrhosis and some important information for gene targeting therapy.
Collapse
Affiliation(s)
- Xia Li
- Transplantation Center, Engineering & Technology Research Center for Transplantation Medicine of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Qiang Wang
- Transplantation Center, Engineering & Technology Research Center for Transplantation Medicine of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Liang Ai
- Transplantation Center, Engineering & Technology Research Center for Transplantation Medicine of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Ke Cheng
- Transplantation Center, Engineering & Technology Research Center for Transplantation Medicine of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| |
Collapse
|
5
|
Mohammadzadeh P, Amberg GC. AXL/Gas6 signaling mechanisms in the hypothalamic-pituitary-gonadal axis. Front Endocrinol (Lausanne) 2023; 14:1212104. [PMID: 37396176 PMCID: PMC10310921 DOI: 10.3389/fendo.2023.1212104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/06/2023] [Indexed: 07/04/2023] Open
Abstract
AXL is a receptor tyrosine kinase commonly associated with a variety of human cancers. Along with its ligand Gas6 (growth arrest-specific protein 6), AXL is emerging as an important regulator of neuroendocrine development and function. AXL signaling in response to Gas6 binding impacts neuroendocrine structure and function at the level of the brain, pituitary, and gonads. During development, AXL has been identified as an upstream inhibitor of gonadotropin receptor hormone (GnRH) production and also plays a key role in the migration of GnRH neurons from the olfactory placode to the forebrain. AXL is implicated in reproductive diseases including some forms of idiopathic hypogonadotropic hypogonadism and evidence suggests that AXL is required for normal spermatogenesis. Here, we highlight research describing AXL/Gas6 signaling mechanisms with a focus on the molecular pathways related to neuroendocrine function in health and disease. In doing so, we aim to present a concise account of known AXL/Gas6 signaling mechanisms to identify current knowledge gaps and inspire future research.
Collapse
|
6
|
Plasma GAS6 predicts mortality risk in acute heart failure patients: insights from the DRAGON-HF trial. J Transl Med 2023; 21:21. [PMID: 36635690 PMCID: PMC9838057 DOI: 10.1186/s12967-022-03859-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/28/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Growth arrest-specific 6 (GAS6) is a vitamin K-dependent protein related to inflammation, fibrosis, as well as platelet function. Genetic ablation of GAS6 in mice protects against cardiac hypertrophy and dysfunction. Nonetheless, the association between plasma GAS6 levels and acute heart failure (AHF) patients is still unknown. METHODS We measured plasma GAS6 concentrations in 1039 patients with AHF who were enrolled in the DRAGON-HF trial (NCT03727828). Mean follow-up of the study was 889 days. The primary endpoint is all-cause death. RESULTS In total, there were 195 primary endpoints of all-cause death and 135 secondary endpoints of cardiovascular death during the mean follow-up duration of 889 days. The higher levels of GAS6 were associated with higher rates of all-cause and cardiovascular death (P < 0.05). Baseline plasma GAS6 levels were still strongly correlated with clinical outcomes in different models after adjustment for clinical factors and N-terminal pro-brain natriuretic peptide (NT-proBNP, P < 0.05). GAS6 could further distinguish the risks of clinical outcomes based on NT-proBNP measurement. CONCLUSION Elevated plasma GAS6 levels were associated with an increased risk of all-cause and cardiovascular death in patients with AHF. Trial registration NCT03727828 (DRAGON-HF trial) clinicaltrials.gov.
Collapse
|
7
|
Rizzi M, Tonello S, D’Onghia D, Sainaghi PP. Gas6/TAM Axis Involvement in Modulating Inflammation and Fibrosis in COVID-19 Patients. Int J Mol Sci 2023; 24:ijms24020951. [PMID: 36674471 PMCID: PMC9861142 DOI: 10.3390/ijms24020951] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Gas6 (growth arrest-specific gene 6) is a widely expressed vitamin K-dependent protein that is involved in many biological processes such as homeostatic regulation, inflammation and repair/fibrotic processes. It is known that it is the main ligand of TAMs, a tyrosine kinase receptor family of three members, namely MerTK, Tyro-3 and Axl, for which it displays the highest affinity. Gas6/TAM axis activation is known to be involved in modulating inflammatory responses as well as fibrotic evolution in many different pathological conditions. Due to the rapidly evolving COVID-19 pandemic, this review will focus on Gas6/TAM axis activation in SARS-CoV-2 infection, where de-regulated inflammatory responses and fibrosis represent a relevant feature of severe disease manifestation. Furthermore, this review will highlight the most recent scientific evidence supporting an unsuspected role of Axl as a SARS-CoV-2 infection driver, and the potential therapeutic advantages of the use of existing Axl inhibitors in COVID-19 management. From a physiological point of view, the Gas6/TAM axis plays a dual role, fostering the tissue repair processes or leading to organ damage and loss of function, depending on the prevalence of its anti-inflammatory or profibrotic properties. This review makes a strong case for further research focusing on the Gas6/TAM axis as a pharmacological target to manage different disease conditions, such as chronic fibrosis or COVID-19.
Collapse
|
8
|
Ortmayr G, Brunnthaler L, Pereyra D, Huber H, Santol J, Rumpf B, Najarnia S, Smoot R, Ammon D, Sorz T, Fritsch F, Schodl M, Voill-Glaninger A, Weitmayr B, Födinger M, Klimpfinger M, Gruenberger T, Assinger A, Mikulits W, Starlinger P. Immunological Aspects of AXL/GAS-6 in the Context of Human Liver Regeneration. Hepatol Commun 2022; 6:576-592. [PMID: 34951136 PMCID: PMC8870037 DOI: 10.1002/hep4.1832] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 08/03/2021] [Accepted: 08/22/2021] [Indexed: 12/13/2022] Open
Abstract
AXL and its corresponding ligand growth arrest-specific 6 (GAS-6) are critically involved in hepatic immunomodulation and regenerative processes. Pleiotropic inhibitory effects on innate inflammatory responses might essentially involve the shift of macrophage phenotype from a pro-inflammatory M1 to an anti-inflammatory M2. We aimed to assess the relevance of the AXL/GAS-6-pathway in human liver regeneration and, consequently, its association with clinical outcome after hepatic resection. Soluble AXL (sAXL) and GAS-6 levels were analyzed at preoperative and postoperative stages in 154 patients undergoing partial hepatectomy and correlated with clinical outcome. Perioperative dynamics of interleukin (IL)-6, soluble tyrosine-protein kinase MER (sMerTK), soluble CD163 (sCD163), and cytokeratin (CK) 18 were assessed to reflect pathophysiological processes. Preoperatively elevated sAXL and GAS-6 levels predicted postoperative liver dysfunction (area under the curve = 0.721 and 0.722; P < 0.005) and worse clinical outcome. These patients failed to respond with an immediate increase of sAXL and GAS-6 upon induction of liver regeneration. Abolished AXL pathway response resulted in a restricted increase of sCD163, suggesting a disrupted phenotypical switch to regeneratory M2 macrophages. No association with sMerTK was observed. Concomitantly, a distinct association of IL-6 levels with an absent increase of AXL/GAS-6 signaling indicated pronounced postoperative inflammation. This was further supported by increased intrahepatic secondary necrosis as reflected by CK18M65. sAXL and GAS-6 represent not only potent and easily accessible preoperative biomarkers for the postoperative outcome but also AXL/GAS-6 signaling might be of critical relevance in human liver regeneration. Refractory AXL/GAS-6 signaling, due to chronic overactivation/stimulation in the context of underlying liver disease, appears to abolish their immediate release following induction of liver regeneration, causing overwhelming immune activation, presumably via intrahepatic immune regulation.
Collapse
Affiliation(s)
- Gregor Ortmayr
- Department of SurgeryMedical University of ViennaGeneral HospitalViennaAustria
| | - Laura Brunnthaler
- Center of Physiology and PharmacologyInstitute of Vascular Biology and Thrombosis ResearchMedical University of ViennaViennaAustria
| | - David Pereyra
- Department of SurgeryMedical University of ViennaGeneral HospitalViennaAustria.,Center of Physiology and PharmacologyInstitute of Vascular Biology and Thrombosis ResearchMedical University of ViennaViennaAustria
| | - Heidemarie Huber
- Department of Medicine IInstitute of Cancer ResearchComprehensive Cancer CenterMedical University of ViennaViennaAustria
| | - Jonas Santol
- Department of SurgeryMedical University of ViennaGeneral HospitalViennaAustria
| | - Benedikt Rumpf
- Department of SurgeryMedical University of ViennaGeneral HospitalViennaAustria
| | - Sina Najarnia
- Department of SurgeryMedical University of ViennaGeneral HospitalViennaAustria
| | - Rory Smoot
- Department of SurgeryMayo ClinicRochesterMNUSA
| | - Daphni Ammon
- Department of SurgeryMedical University of ViennaGeneral HospitalViennaAustria
| | - Thomas Sorz
- Department of SurgeryMedical University of ViennaGeneral HospitalViennaAustria
| | - Fabian Fritsch
- Department of SurgeryMedical University of ViennaGeneral HospitalViennaAustria
| | - Michael Schodl
- Department of SurgeryMedical University of ViennaGeneral HospitalViennaAustria
| | - Astrid Voill-Glaninger
- Department of Laboratory MedicineViennese Health Network, Clinic LandstraßeViennaAustria
| | - Barbara Weitmayr
- Department of PathologyViennese Health Network, Clinic LandstraßeViennaAustria
| | - Manuela Födinger
- Department of Laboratory MedicineViennese Health NetworkClinic FavoritenViennaAustria
| | - Martin Klimpfinger
- Department of PathologyViennese Health NetworkClinic FavoritenViennaAustria
| | - Thomas Gruenberger
- Department of SurgeryHPB Center, Viennese Health Network, Clinic Favoriten and Sigmund Freud Private UniversityViennaAustria
| | - Alice Assinger
- Center of Physiology and PharmacologyInstitute of Vascular Biology and Thrombosis ResearchMedical University of ViennaViennaAustria
| | - Wolfgang Mikulits
- Department of Medicine IInstitute of Cancer ResearchComprehensive Cancer CenterMedical University of ViennaViennaAustria
| | - Patrick Starlinger
- Department of SurgeryMedical University of ViennaGeneral HospitalViennaAustria.,Department of SurgeryMayo ClinicRochesterMNUSA
| |
Collapse
|
9
|
Smirne C, Croce E, Di Benedetto D, Cantaluppi V, Comi C, Sainaghi PP, Minisini R, Grossini E, Pirisi M. Oxidative Stress in Non-Alcoholic Fatty Liver Disease. LIVERS 2022; 2:30-76. [DOI: 10.3390/livers2010003] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a challenging disease caused by multiple factors, which may partly explain why it still remains an orphan of adequate therapies. This review highlights the interaction between oxidative stress (OS) and disturbed lipid metabolism. Several reactive oxygen species generators, including those produced in the gastrointestinal tract, contribute to the lipotoxic hepatic (and extrahepatic) damage by fatty acids and a great variety of their biologically active metabolites in a “multiple parallel-hit model”. This leads to inflammation and fibrogenesis and contributes to NAFLD progression. The alterations of the oxidant/antioxidant balance affect also metabolism-related organelles, leading to lipid peroxidation, mitochondrial dysfunction, and endoplasmic reticulum stress. This OS-induced damage is at least partially counteracted by the physiological antioxidant response. Therefore, modulation of this defense system emerges as an interesting target to prevent NAFLD development and progression. For instance, probiotics, prebiotics, diet, and fecal microbiota transplantation represent new therapeutic approaches targeting the gut microbiota dysbiosis. The OS and its counter-regulation are under the influence of individual genetic and epigenetic factors as well. In the near future, precision medicine taking into consideration genetic or environmental epigenetic risk factors, coupled with new OS biomarkers, will likely assist in noninvasive diagnosis and monitoring of NAFLD progression and in further personalizing treatments.
Collapse
Affiliation(s)
- Carlo Smirne
- Department of Translational Medicine, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Eleonora Croce
- Department of Translational Medicine, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Davide Di Benedetto
- Department of Translational Medicine, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Vincenzo Cantaluppi
- Department of Translational Medicine, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Cristoforo Comi
- Department of Translational Medicine, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Pier Paolo Sainaghi
- Department of Translational Medicine, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Rosalba Minisini
- Department of Translational Medicine, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Elena Grossini
- Department of Translational Medicine, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Mario Pirisi
- Department of Translational Medicine, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| |
Collapse
|
10
|
Abstract
TAM receptors (Tyro3, Axl and MerTK) are a family of tyrosine kinase receptors that are expressed in a variety of cell populations, including liver parenchymal and non-parenchymal cells. These receptors are vital for immune homeostasis, as they regulate the innate immune response by suppressing inflammation via toll-like receptor inhibition and by promoting tissue resolution through efferocytosis. However, there is increasing evidence indicating that aberrant TAM receptor signaling may play a role in pathophysiological processes in the context of liver disease. This review will explore the roles of TAM receptors and their ligands in liver homeostasis as well as a variety of disease settings, including acute liver injury, steatosis, fibrosis, cirrhosis-associated immune dysfunction and hepatocellular carcinoma. A better understanding of our current knowledge of TAM receptors in liver disease may identify new opportunities for disease monitoring as well as novel therapeutic targets. Nonetheless, this review also aims to highlight areas where further research on TAM receptor biology in liver disease is required.
Collapse
|
11
|
Bhave S, Ho HK. Exploring the Gamut of Receptor Tyrosine Kinases for Their Promise in the Management of Non-Alcoholic Fatty Liver Disease. Biomedicines 2021; 9:1776. [PMID: 34944593 PMCID: PMC8698495 DOI: 10.3390/biomedicines9121776] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 11/17/2022] Open
Abstract
Recently, non-alcoholic fatty liver disease (NAFLD) has emerged as a predominant health concern affecting approximately a quarter of the world's population. NAFLD is a spectrum of liver ailments arising from nascent lipid accumulation and leading to inflammation, fibrosis or even carcinogenesis. Despite its prevalence and severity, no targeted pharmacological intervention is approved to date. Thus, it is imperative to identify suitable drug targets critical to the development and progression of NAFLD. In this quest, a ray of hope is nestled within a group of proteins, receptor tyrosine kinases (RTKs), as targets to contain or even reverse NAFLD. RTKs control numerous vital biological processes and their selective expression and activity in specific diseases have rendered them useful as drug targets. In this review, we discuss the recent advancements in characterizing the role of RTKs in NAFLD progression and qualify their suitability as pharmacological targets. Available data suggests inhibition of Epidermal Growth Factor Receptor, AXL, Fibroblast Growth Factor Receptor 4 and Vascular Endothelial Growth Factor Receptor, and activation of cellular mesenchymal-epithelial transition factor and Fibroblast Growth Factor Receptor 1 could pave the way for novel NAFLD therapeutics. Thus, it is important to characterize these RTKs for target validation and proof-of-concept through clinical trials.
Collapse
Affiliation(s)
| | - Han Kiat Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117559, Singapore;
| |
Collapse
|
12
|
Zhang J, Liu Q, He J, Li Y. Novel Therapeutic Targets in Liver Fibrosis. Front Mol Biosci 2021; 8:766855. [PMID: 34805276 PMCID: PMC8602792 DOI: 10.3389/fmolb.2021.766855] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/18/2021] [Indexed: 02/05/2023] Open
Abstract
Liver fibrosis is end-stage liver disease that can be rescued. If irritation continues due to viral infection, schistosomiasis and alcoholism, liver fibrosis can progress to liver cirrhosis and even cancer. The US Food and Drug Administration has not approved any drugs that act directly against liver fibrosis. The only treatments currently available are drugs that eliminate pathogenic factors, which show poor efficacy; and liver transplantation, which is expensive. This highlights the importance of clarifying the mechanism of liver fibrosis and searching for new treatments against it. This review summarizes how parenchymal, nonparenchymal cells, inflammatory cells and various processes (liver fibrosis, hepatic stellate cell activation, cell death and proliferation, deposition of extracellular matrix, cell metabolism, inflammation and epigenetics) contribute to liver fibrosis. We highlight discoveries of novel therapeutic targets, which may provide new insights into potential treatments for liver fibrosis.
Collapse
Affiliation(s)
- Jinhang Zhang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Sichuan, China
| | - Qinhui Liu
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Sichuan, China
| | - Jinhan He
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Sichuan, China.,Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, China
| | - Yanping Li
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Sichuan, China
| |
Collapse
|
13
|
Liebold I, Jawazneh AA, Hamley M, Bosurgi L. Apoptotic cell signals and heterogeneity in macrophage function: Fine-tuning for a healthy liver. Semin Cell Dev Biol 2021; 119:72-81. [PMID: 34246569 DOI: 10.1016/j.semcdb.2021.06.012] [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: 12/22/2020] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 12/13/2022]
Abstract
Functional heterogeneity in tissue macrophage populations has often been traced to developmental and spatial cues. Upon tissue damage, macrophages are exposed to soluble mediators secreted by activated cells, which shape their polarisation. Interestingly, macrophages are concomitantly exposed to a variety of different dying cells, which carry miscellaneous signals and that need to be recognised and promptly up-taken by professional phagocytes. This review discusses how differences in the nature of the dying cells, like their morphological and biochemical features as well as the specificity of phagocytic receptor usage on macrophages, might contribute to the transcriptional and functional heterogeneity observed in phagocytic cells in the tissue.
Collapse
Affiliation(s)
- Imke Liebold
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Amirah Al Jawazneh
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Madeleine Hamley
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Lidia Bosurgi
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
| |
Collapse
|
14
|
Yılmaz Y, Batur T, Korhan P, Öztürk M, Atabey N. Targeting c-Met and AXL Crosstalk for the Treatment of Hepatocellular Carcinoma. LIVER CANCER IN THE MIDDLE EAST 2021:333-364. [DOI: 10.1007/978-3-030-78737-0_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
15
|
Hayashi M, Abe K, Fujita M, Takahashi A, Hashimoto Y, Ohira H. Serum Gas6 and Axl as non-invasive biomarkers of advanced histological stage in primary biliary cholangitis. Hepatol Res 2020; 50:1337-1346. [PMID: 32885557 DOI: 10.1111/hepr.13568] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 08/10/2020] [Accepted: 08/23/2020] [Indexed: 12/13/2022]
Abstract
AIM Advanced histological stage is an important factor in individual risk stratification in patients with primary biliary cholangitis (PBC). Non-invasive biomarkers for advanced histological stage are needed. We assessed the utility of Gas6 and Axl as biomarkers for advanced histological stage in patients with PBC. METHODS A total of 113 biopsy-proven PBC patients and 20 healthy controls were included in this study. Serum Axl and Gas6 were measured using enzyme-linked immunosorbent assay. The Gas6 / albumin ratio and Axl / albumin ratio were also evaluated as biomarkers of histological stage. RESULTS Serum Axl (42.6 ng/mL vs. 30.6 ng/mL, P < 0.001) and Gas6 (21.1 ng/mL vs. 18.8 ng/mL, P = 0.007) levels in PBC patients were significantly higher than those in healthy controls. The Axl / albumin ratio was 10.4, and the Gas6 / albumin ratio was 7.6 in patients with PBC. Gas6 and Axl were significantly correlated with aspartate aminotransferase, bilirubin, albumin, and platelets. Gas6 and Axl levels in patients with an advanced Scheuer stage and an advanced Nakanuma stage were significantly higher than those in other patients. The area under the receiver operating characteristic curve (AUROC) of Axl, Gas6, Axl / albumin, and Gas6 / albumin for diagnosing Scheuer stage 4 was 0.733, 0.837, 0.845, and 0.893, respectively. The AUROC of Axl, Gas6, Axl / albumin, and Gas6 / albumin for diagnosing Nakanuma stage 4 was 0.794, 0.834, 0.869, and 0.898, respectively. CONCLUSION High levels of Gas6 and Axl were associated with advanced histological stage in PBC patients. Furthermore, the Gas6 / albumin ratio and the Axl / albumin ratio showed a high AUROC for diagnosing advanced histological stage.
Collapse
Affiliation(s)
- Manabu Hayashi
- Department of Gastroenterology, Fukushima Medical University, Fukushima, Japan
| | - Kazumichi Abe
- Department of Gastroenterology, Fukushima Medical University, Fukushima, Japan
| | - Masashi Fujita
- Department of Gastroenterology, Fukushima Medical University, Fukushima, Japan
| | - Atsushi Takahashi
- Department of Gastroenterology, Fukushima Medical University, Fukushima, Japan
| | - Yuko Hashimoto
- Department of Diagnostic Pathology, Fukushima Medical University, Fukushima, Japan
| | - Hiromasa Ohira
- Department of Gastroenterology, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
16
|
DuBois JC, Ray AK, Davies P, Shafit-Zagardo B. Anti-Axl antibody treatment reduces the severity of experimental autoimmune encephalomyelitis. J Neuroinflammation 2020; 17:324. [PMID: 33121494 PMCID: PMC7599105 DOI: 10.1186/s12974-020-01982-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Multiple sclerosis is an immune-mediated disease of the central nervous system (CNS) characterized by inflammation, oligodendrocytes loss, demyelination, and damaged axons. Tyro3, Axl, and MerTK belong to a family of receptor tyrosine kinases that regulate innate immune responses and CNS homeostasis. During experimental autoimmune encephalomyelitis (EAE), the mRNA expression of MerTK, Gas6, and Axl significantly increase, whereas Tyro3 and ProS1 remain unchanged. We have shown that Gas6 is neuroprotective during EAE, and since Gas6 activation of Axl may be necessary for conferring neuroprotection, we sought to determine whether α-Axl or α-MerTK antibodies, shown by others to activate their respective receptors in vivo, could effectively reduce inflammation and neurodegeneration. METHODS Mice received either α-Axl, α-MerTK, IgG isotype control, or PBS before the onset of EAE symptoms. EAE clinical course, axonal damage, demyelination, cytokine production, and immune cell activation in the CNS were used to determine the severity of EAE. RESULTS α-Axl antibody treatment significantly decreased the EAE clinical indices of female mice during chronic EAE and of male mice during both acute and chronic phases. The number of days mice were severely paralyzed also significantly decreased with α-Axl treatment. Inflammatory macrophages/microglia and the extent of demyelination significantly decreased in the spinal cords of α-Axl-treated mice during chronic EAE, with no differences in the production of pro-inflammatory cytokines. α-MerTK antibody did not influence EAE induction or progression. CONCLUSION Our data suggests that the beneficial effect of Gas6/Axl signaling observed in mice administered with Gas6 can be partially preserved by administering an activating α-Axl antibody, but not α-MerTK.
Collapse
Affiliation(s)
- Juwen C. DuBois
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461 USA
| | - Alex K. Ray
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY USA
| | - Peter Davies
- North Shore-LIJ Health System, Feinstein Institute for Medical Research, Manhasset, NY USA
| | - Bridget Shafit-Zagardo
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461 USA
| |
Collapse
|
17
|
Wan S, Luo F, Huang C, Liu C, Luo Q, Zhu X. Ursolic acid reverses liver fibrosis by inhibiting interactive NOX4/ROS and RhoA/ROCK1 signalling pathways. Aging (Albany NY) 2020; 12:10614-10632. [PMID: 32496208 PMCID: PMC7346053 DOI: 10.18632/aging.103282] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023]
Abstract
Liver fibrosis is the reversible deposition of extracellular matrix (ECM) and scar formation after liver damage by various stimuli. The interaction between NOX4/ROS and RhoA/ROCK1 in liver fibrosis is not yet clear. Ursolic acid (UA) is a traditional Chinese medicine with anti-fibrotic effects, but the molecular mechanism underlying these effects is still unclear. We investigated the interaction between NOX4/ROS and RhoA/ROCK1 during liver fibrosis and whether these molecules are targets for the anti-fibrotic effects of UA. First, we confirmed that UA reversed CCl4-induced liver fibrosis. In the NOX4 intervention and RhoA intervention groups, related experimental analyses confirmed the decrease in CCl4-induced liver fibrosis. Next, we determined that the expression of NOX4 and RhoA/ROCK1 was decreased in UA-treated liver fibrotic mice. Furthermore, RhoA/ROCK1 expression was decreased in the NOX4 intervention group, but there was no significant change in the expression of NOX4 in the RhoA intervention group. Finally, we found that liver fibrotic mice showed a decline in their microbiota diversity and abundance, a change in their microbiota composition, and a reduction in the number of potential beneficial bacteria. However, in UA-treated liver fibrotic mice, the microbiota dysbiosis was ameliorated. In conclusion, the NOX4/ROS and RhoA/ROCK1 signalling pathways are closely linked to the development of liver fibrosis. UA can reverse liver fibrosis by inhibiting the NOX4/ROS and RhoA/ROCK1 signalling pathways, which may interact with each other.
Collapse
Affiliation(s)
- Sizhe Wan
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Fangyun Luo
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Chenkai Huang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Cong Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qingtian Luo
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xuan Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
18
|
Role of Gas6 and TAM Receptors in the Identification of Cardiopulmonary Involvement in Systemic Sclerosis and Scleroderma Spectrum Disorders. DISEASE MARKERS 2020; 2020:2696173. [PMID: 32454903 PMCID: PMC7240795 DOI: 10.1155/2020/2696173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 04/24/2020] [Indexed: 01/25/2023]
Abstract
Background Few biomarkers are available for early identification of pulmonary arterial hypertension (PAH) and interstitial lung disease (ILD) in systemic sclerosis (SS) and scleroderma spectrum disorders (SSD). Aims To evaluate Gas6, sAxl, and sMer as biomarkers for cardiopulmonary complications of SS and SSD. Methods In a cross-sectional observational study, we recruited 125 consecutive patients, affected by SS and SSD and referred to a tertiary-level pulmonary hypertension outpatient clinic. All patients underwent a comprehensive evaluation for identification of PAH and ILD. Gas6, sMer, and sAxl concentrations were measured with ELISA protocols, and concentrations were compared according to PAH or ILD. Results Nineteen subjects had pulmonary hypertension (PH) (14 PAH), and 39 had ILD (6 severe). Plasma sMer was increased in PAH (18.6 ng/ml IQR [11.7-20.3]) with respect to the absence (12.4 [8.0-15.8]) or other form of pulmonary hypertension (9.6 [7.4-12.5]; K–W variance p < 0.04). Conversely, Gas6 and sAxl levels were slightly increased in mild ILD (25.8 ng/ml [19.5-32.1] and 24.6 [20.1-32.5]) and reduced in severe ILD (16.6 [15.0-22.1] and 15.5 [14.9-22.4]) in comparison to no evidence of ILD (23.4 [18.8-28.1] and 21.6 [18.1-28.4]; K–W, p ≤ 0.05). Plasma sMer ≥ 19 ng/ml has 50% sensitivity and 92% specificity in PAH identification (area under the ROC curve (AUC) 0.697, p < 0.03). Values of Gas6 ≤ 24.5 ng/ml and of sAxl ≤ 15.5 ng/ml have 100% and 67% sensitivity and 47% and 86% specificity, respectively, in identifying severe ILD (Gas6 AUC 0.787, p < 0.001; sAxl AUC 0.705, p < 0.05). Conclusions The assay of Gas6 sAxl and sMer may be useful to help in the identification of PAH and ILD in SS and SSD patients. The Gas6/TAM system seems to be relevant in cardiopulmonary complications of SS and SSD and merits further investigations.
Collapse
|
19
|
Zhao M, Jung Y, Jiang Z, Svensson KJ. Regulation of Energy Metabolism by Receptor Tyrosine Kinase Ligands. Front Physiol 2020; 11:354. [PMID: 32372975 PMCID: PMC7186430 DOI: 10.3389/fphys.2020.00354] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 03/26/2020] [Indexed: 12/14/2022] Open
Abstract
Metabolic diseases, such as diabetes, obesity, and fatty liver disease, have now reached epidemic proportions. Receptor tyrosine kinases (RTKs) are a family of cell surface receptors responding to growth factors, hormones, and cytokines to mediate a diverse set of fundamental cellular and metabolic signaling pathways. These ligands signal by endocrine, paracrine, or autocrine means in peripheral organs and in the central nervous system to control cellular and tissue-specific metabolic processes. Interestingly, the expression of many RTKs and their ligands are controlled by changes in metabolic demand, for example, during starvation, feeding, or obesity. In addition, studies of RTKs and their ligands in regulating energy homeostasis have revealed unexpected diversity in the mechanisms of action and their specific metabolic functions. Our current understanding of the molecular, biochemical and genetic control of energy homeostasis by the endocrine RTK ligands insulin, FGF21 and FGF19 are now relatively well understood. In addition to these classical endocrine signals, non-endocrine ligands can govern local energy regulation, and the intriguing crosstalk between the RTK family and the TGFβ receptor family demonstrates a signaling network that diversifies metabolic process between tissues. Thus, there is a need to increase our molecular and mechanistic understanding of signal diversification of RTK actions in metabolic disease. Here we review the known and emerging molecular mechanisms of RTK signaling that regulate systemic glucose and lipid metabolism, as well as highlighting unexpected roles of non-classical RTK ligands that crosstalk with other receptor pathways.
Collapse
Affiliation(s)
- Meng Zhao
- Department of Pathology, Stanford University, Stanford, CA, United States.,Stanford Diabetes Research Center, Stanford, CA, United States
| | - Yunshin Jung
- Department of Pathology, Stanford University, Stanford, CA, United States.,Stanford Diabetes Research Center, Stanford, CA, United States
| | - Zewen Jiang
- Department of Pathology, Stanford University, Stanford, CA, United States.,Stanford Diabetes Research Center, Stanford, CA, United States
| | - Katrin J Svensson
- Department of Pathology, Stanford University, Stanford, CA, United States.,Stanford Diabetes Research Center, Stanford, CA, United States
| |
Collapse
|
20
|
Wang Q, Zhao Y, Zang B. Anti-inflammation and anti-apoptosis effects of growth arrest-specific protein 6 in acute liver injury induced by LPS/D-GalN in mice. Acta Cir Bras 2020; 35:e202000204. [PMID: 32294688 PMCID: PMC7158606 DOI: 10.1590/s0102-865020200020000004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/16/2020] [Indexed: 02/07/2023] Open
Abstract
Purpose To investigate the effect of growth arrest-specific protein 6 (Gas6) on acute liver injury in mice and related mechanisms. Methods Thirty C57BL/6 (6-8 weeks old) mice were randomly divided into control, LPS/D-GalN, and LPS/D-GalN+Gas6 groups (10 mice in each group). The LPS/D-GalN group was intraperitoneally administered with LPS (0.25 mg/Kg) and D-GalN (400 mg/Kg) for 5h. The LPS/D-GalN+Gas6 group was intraperitoneally administered with rmGas6 one hour before intraperitoneal application of LPS/D-GalN. All subjects were sacrificed at 5 h for blood and tissue analysis. The expression of protein and mRNA was assessed by western blotting and RT-PCR, respectively. Results Compared with the control group, AST, ALT, IL-1β, TNF-α, IL-6 IL-10, MPO activity were increased in the LPS/D-GalN group. However, they were significantly inhibited by Gas6. Gas6 markedly suppressed the expression of apoptosis-related protein induced by LPS/D-GalN. Moreover, Gas6 attenuated the activation of the NF-κB signaling pathway in acute liver injury induced by LPS/D-GalN. Conclusions Gas6 alleviates acute liver injury in mice through regulating NF-κB signaling pathways. Gas6 can be a potential therapeutic agent in treating LPS/D-GalN-induced acute liver injury in the future.
Collapse
Affiliation(s)
- Qian Wang
- China Medical University, China; China Medical University, China
| | | | | |
Collapse
|
21
|
Cai B, Dongiovanni P, Corey KE, Wang X, Shmarakov IO, Zheng Z, Kasikara C, Davra V, Meroni M, Chung RT, Rothlin CV, Schwabe RF, Blaner WS, Birge RB, Valenti L, Tabas I. Macrophage MerTK Promotes Liver Fibrosis in Nonalcoholic Steatohepatitis. Cell Metab 2020; 31:406-421.e7. [PMID: 31839486 PMCID: PMC7004886 DOI: 10.1016/j.cmet.2019.11.013] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 10/07/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is emerging as a leading cause of chronic liver disease. However, therapeutic options are limited by incomplete understanding of the mechanisms of NASH fibrosis, which is mediated by activation of hepatic stellate cells (HSCs). In humans, human genetic studies have shown that hypomorphic variations in MERTK, encoding the macrophage c-mer tyrosine kinase (MerTK) receptor, provide protection against liver fibrosis, but the mechanisms remain unknown. We now show that holo- or myeloid-specific Mertk targeting in NASH mice decreases liver fibrosis, congruent with the human genetic data. Furthermore, ADAM metallopeptidase domain 17 (ADAM17)-mediated MerTK cleavage in liver macrophages decreases during steatosis to NASH transition, and mice with a cleavage-resistant MerTK mutant have increased NASH fibrosis. Macrophage MerTK promotes an ERK-TGFβ1 pathway that activates HSCs and induces liver fibrosis. These data provide insights into the role of liver macrophages in NASH fibrosis and provide a plausible mechanism underlying MERTK as a genetic risk factor for NASH fibrosis.
Collapse
Affiliation(s)
- Bishuang Cai
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Paola Dongiovanni
- General Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Kathleen E Corey
- Liver Center, Gastrointestinal Division, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Igor O Shmarakov
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ze Zheng
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Canan Kasikara
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Viralkumar Davra
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University, New Jersey Medical School Cancer Center, Newark, NJ 07103, USA
| | - Marica Meroni
- General Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Raymond T Chung
- Liver Center, Gastrointestinal Division, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA, USA
| | - Carla V Rothlin
- Department of Immunobiology, Yale University School of Medicine and Department of Pharmacology, Yale University, New Haven, CT, USA
| | - Robert F Schwabe
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - William S Blaner
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Raymond B Birge
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University, New Jersey Medical School Cancer Center, Newark, NJ 07103, USA
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milano 20122, Italy; Translational Medicine - Transfusion Medicine and Hematology, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Departments of Pathology & Cell Biology and Physiology & Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
22
|
Cubero FJ. Staging NAFLD: Diagnostic and Therapeutic Value of TAM Signaling. Cell Mol Gastroenterol Hepatol 2019; 9:545-546. [PMID: 31881182 PMCID: PMC7078440 DOI: 10.1016/j.jcmgh.2019.11.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/10/2022]
Affiliation(s)
- Francisco Javier Cubero
- Correspondence Address correspondence to: Francisco Javier Cubero, PhD, Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, c/Doctor Severo Ochoa, 9, Madrid 28040, Spain.
| |
Collapse
|
23
|
Tutusaus A, de Gregorio E, Cucarull B, Cristóbal H, Aresté C, Graupera I, Coll M, Colell A, Gausdal G, Lorens JB, García de Frutos P, Morales A, Marí M. A Functional Role of GAS6/TAM in Nonalcoholic Steatohepatitis Progression Implicates AXL as Therapeutic Target. Cell Mol Gastroenterol Hepatol 2019; 9:349-368. [PMID: 31689560 PMCID: PMC7013198 DOI: 10.1016/j.jcmgh.2019.10.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS GAS6 signaling, through the TAM receptor tyrosine kinases AXL and MERTK, participates in chronic liver pathologies. Here, we addressed GAS6/TAM involvement in Non-Alcoholic SteatoHepatitis (NASH) development. METHODS GAS6/TAM signaling was analyzed in cultured primary hepatocytes, hepatic stellate cells (HSC) and Kupffer cells (KCs). Axl-/-, Mertk-/- and wild-type C57BL/6 mice were fed with Chow, High Fat Choline-Deficient Methionine-Restricted (HFD) or methionine-choline-deficient (MCD) diet. HSC activation, liver inflammation and cytokine/chemokine production were measured by qPCR, mRNA Array analysis, western blotting and ELISA. GAS6, soluble AXL (sAXL) and MERTK (sMERTK) levels were analyzed in control individuals, steatotic and NASH patients. RESULTS In primary mouse cultures, GAS6 or MERTK activation protected primary hepatocytes against lipid toxicity via AKT/STAT-3 signaling, while bemcentinib (small molecule AXL inhibitor BGB324) blocked AXL-induced fibrogenesis in primary HSCs and cytokine production in LPS-treated KCs. Accordingly; bemcentinib diminished liver inflammation and fibrosis in MCD- and HFD-fed mice. Upregulation of AXL and ADAM10/ADAM17 metalloproteinases increased sAXL in HFD-fed mice. Transcriptome profiling revealed major reduction in fibrotic- and inflammatory-related genes in HFD-fed mice after bemcentinib administration. HFD-fed Mertk-/- mice exhibited enhanced NASH, while Axl-/- mice were partially protected. In human serum, sAXL levels augmented even at initial stages, whereas GAS6 and sMERTK increased only in cirrhotic NASH patients. In agreement, sAXL increased in HFD-fed mice before fibrosis establishment, while bemcentinib prevented liver fibrosis/inflammation in early NASH. CONCLUSION AXL signaling, increased in NASH patients, promotes fibrosis in HSCs and inflammation in KCs, while GAS6 protects cultured hepatocytes against lipotoxicity via MERTK. Bemcentinib, by blocking AXL signaling and increasing GAS6 levels, reduces experimental NASH, revealing AXL as an effective therapeutic target for clinical practice.
Collapse
Affiliation(s)
- Anna Tutusaus
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain,Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Estefanía de Gregorio
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain
| | - Blanca Cucarull
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain,Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Helena Cristóbal
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain
| | - Cristina Aresté
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain
| | - Isabel Graupera
- Liver Unit, Hospital Clínic, Biomedical Research Networking Center in Hepatic and Digestive Diseases, Barcelona, Spain
| | - Mar Coll
- Liver Unit, Hospital Clínic, Biomedical Research Networking Center in Hepatic and Digestive Diseases, Barcelona, Spain
| | - Anna Colell
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain
| | | | - James B. Lorens
- BerGenBio AS, Bergen, Norway,Department of Biomedicine, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Pablo García de Frutos
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain,Correspondence Address correspondence to: Montserrat Marí, PhD, Albert Morales, PhD, or Pablo García de Frutos, PhD, Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), C/ Rosselló 161, 6th Floor, 08036 Barcelona, Spain. fax: +34-93-3638301.
| | - Albert Morales
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain,Barcelona Clinic Liver Cancer Group, Liver Unit, Hospital Clínic, Biomedical Research Networking Center in Hepatic and Digestive Diseases, Barcelona, Spain,Correspondence Address correspondence to: Montserrat Marí, PhD, Albert Morales, PhD, or Pablo García de Frutos, PhD, Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), C/ Rosselló 161, 6th Floor, 08036 Barcelona, Spain. fax: +34-93-3638301.
| | - Montserrat Marí
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain,Correspondence Address correspondence to: Montserrat Marí, PhD, Albert Morales, PhD, or Pablo García de Frutos, PhD, Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), C/ Rosselló 161, 6th Floor, 08036 Barcelona, Spain. fax: +34-93-3638301.
| |
Collapse
|
24
|
Gas6/TAM System: A Key Modulator of the Interplay between Inflammation and Fibrosis. Int J Mol Sci 2019; 20:ijms20205070. [PMID: 31614787 PMCID: PMC6834320 DOI: 10.3390/ijms20205070] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/01/2019] [Accepted: 10/08/2019] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is the result of an overly abundant deposition of extracellular matrix (ECM) due to the fact of repetitive tissue injuries and/or dysregulation of the repair process. Fibrogenesis is a pathogenetic phenomenon which is involved in different chronic human diseases, accounting for a high burden of morbidity and mortality. Despite being triggered by different causative factors, fibrogenesis follows common pathways, the knowledge of which is, however, still unsatisfactory. This represents a significant limit for the development of effective antifibrotic drugs. In the present paper, we aimed to review the current evidence regarding the potential role played in fibrogenesis by growth arrest-specific 6 (Gas6) and its receptors Tyro3 protein tyrosine kinase (Tyro3), Axl receptor tyrosine kinase (Axl), and Mer tyrosine kinase protooncogene (MerTK) (TAM). Moreover, we aimed to review data about the pathogenetic role of this system in the development of different human diseases characterized by fibrosis. Finally, we aimed to explore the potential implications of these findings in diagnosis and treatment.
Collapse
|
25
|
TAM Receptor Pathways at the Crossroads of Neuroinflammation and Neurodegeneration. DISEASE MARKERS 2019; 2019:2387614. [PMID: 31636733 PMCID: PMC6766163 DOI: 10.1155/2019/2387614] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/04/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023]
Abstract
Increasing evidence suggests that pathogenic mechanisms underlying neurodegeneration are strongly linked with neuroinflammatory responses. Tyro3, Axl, and Mertk (TAM receptors) constitute a subgroup of the receptor tyrosine kinase family, cell surface receptors which transmit signals from the extracellular space to the cytoplasm and nucleus. TAM receptors and the corresponding ligands, Growth Arrest Specific 6 and Protein S, are expressed in different tissues, including the nervous system, playing complex roles in tissue repair, inflammation and cell survival, proliferation, and migration. In the nervous system, TAM receptor signalling modulates neurogenesis and neuronal migration, synaptic plasticity, microglial activation, phagocytosis, myelination, and peripheral nerve repair, resulting in potential interest in neuroinflammatory and neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Multiple Sclerosis. In Alzheimer and Parkinson diseases, a role of TAM receptors in neuronal survival and pathological protein aggregate clearance has been suggested, while in Multiple Sclerosis TAM receptors are involved in myelination and demyelination processes. To better clarify roles and pathways involving TAM receptors may have important therapeutic implications, given the fine modulation of multiple molecular processes which could be reached. In this review, we summarise the roles of TAM receptors in the central nervous system, focusing on the regulation of immune responses and microglial activities and analysing in vitro and in vivo studies regarding TAM signalling involvement in neurodegeneration.
Collapse
|
26
|
Gas6/TAM Signaling Components as Novel Biomarkers of Liver Fibrosis. DISEASE MARKERS 2019; 2019:2304931. [PMID: 31583026 PMCID: PMC6754881 DOI: 10.1155/2019/2304931] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/20/2019] [Accepted: 08/12/2019] [Indexed: 12/14/2022]
Abstract
Liver fibrosis consists in the accumulation of extracellular matrix components mainly derived from activated hepatic stellate cells. This is commonly the result of chronic liver injury repair and represents an important health concern. As liver biopsy is burdened with many drawbacks, not surprisingly there is great interest to find new reliable noninvasive methods. Among the many are new potential fibrosis biomarkers under study, some of the most promising represented by the growth arrest-specific gene 6 (Gas6) serum protein and its family of tyrosine kinase receptors, namely, Tyro3, Axl, and MERTK (TAM). Gas6/TAM system (mainly, Axl and MERTK) has in fact recently emerged as an important player in the progression of liver fibrosis. This review is aimed at giving an overall perspective of the roles played by these molecules in major chronic liver diseases. The most promising findings up to date acknowledge that both Gas6 and its receptor serum levels (such as sAxl and, probably, sMERTK) have been shown to potentially allow for easy and accurate measurement of hepatic fibrosis progression, also providing indicative parameters of hepatic dysfunction. Although most of the current scientific evidence is still preliminary and there are no in vivo validation studies on large patient series, it still looks very promising to imagine a possible future prognostic role for these biomarkers in the multidimensional assessment of a liver patient. One may also speculate on a potential role for this system targeting (e.g., with small molecule inhibitors against Axl) as a therapeutic strategy for liver fibrosis management, always bearing in mind that any such therapeutic approach might face toxicity.
Collapse
|
27
|
Axl expression is increased in early stages of left ventricular remodeling in an animal model with pressure-overload. PLoS One 2019; 14:e0217926. [PMID: 31181097 PMCID: PMC6557565 DOI: 10.1371/journal.pone.0217926] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 05/21/2019] [Indexed: 01/24/2023] Open
Abstract
Background AXL is a receptor tyrosine kinase that has been related to kidney and vascular disorders. Heart failure patients with reduced ejection fraction have higher AXL in serum than controls. No information about Axl expression with HF progression is available. Methods Thoracic transverse aortic constriction (TAC) was successfully performed on male Wistar rats (n = 25) with different constriction levels. Controls underwent sham surgery (n = 12). Echocardiography measurements were performed 4–8 weeks after surgery. Collagen deposition was measured with picrosirius red staining. Axl mRNA levels in left ventricle (LV), left kidney (LK) and ascending aorta (aAo) and the LV expression of cardiac remodeling and fibrogenic factors were quantified with real-time PCR. AXL LV protein levels were quantified with western blot and localization was analyzed by immunohistochemistry. Soluble AXL levels in plasma were assayed with ELISA. Results Successful TAC rats were classified into LV hypertrophy (LVH) or heart failure (HF), modeling the progressive cardiac changes after pressure overload. Collagen deposition was increased only in the HF group. LV Axl mRNA levels were higher in LVH and HF than in Sham rats, and correlated with LVHi, and hypertrophic and fibrogenic mediators. However, no association was found with LV systolic function. AXL was expressed in LV myocytes and other cell types. Concentration of circulating sAXL in plasma was increased in the LVH group compared to Sham and HF rats. Axl mRNA levels were similar in all groups in the LK and aAo. Conclusions Axl expression pattern suggests a role in the early progression of LV remodeling in HF but not in the later systolic dysfunction. The higher levels of circulating AXL found in HF patients most probably shed from the heart.
Collapse
|
28
|
High Risk of Hepatocellular Carcinoma Development in Fibrotic Liver: Role of the Hippo-YAP/TAZ Signaling Pathway. Int J Mol Sci 2019; 20:ijms20030581. [PMID: 30700007 PMCID: PMC6387126 DOI: 10.3390/ijms20030581] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023] Open
Abstract
Liver cancer is the fourth leading cause of cancer-related death globally, accounting for approximately 800,000 deaths annually. Hepatocellular carcinoma (HCC) is the most common type of liver cancer, making up about 80% of cases. Liver fibrosis and its end-stage disease, cirrhosis, are major risk factors for HCC. A fibrotic liver typically shows persistent hepatocyte death and compensatory regeneration, chronic inflammation, and an increase in reactive oxygen species, which collaboratively create a tumor-promoting microenvironment via inducing genetic alterations and chromosomal instability, and activating various oncogenic molecular signaling pathways. In this article, we review recent advances in fields of liver fibrosis and carcinogenesis, and consider several molecular signaling pathways that promote hepato-carcinogenesis under the microenvironment of liver fibrosis. In particular, we pay attention to emerging roles of the Hippo-YAP/TAZ signaling pathway in stromal activation, hepatic fibrosis, and liver cancer.
Collapse
|
29
|
Dynamics of Axl Receptor Shedding in Hepatocellular Carcinoma and Its Implication for Theranostics. Int J Mol Sci 2018; 19:ijms19124111. [PMID: 30567378 PMCID: PMC6321118 DOI: 10.3390/ijms19124111] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022] Open
Abstract
Signaling of the receptor tyrosine kinase Axl and its ligand Gas6 is crucially involved in the development of liver fibrosis and hepatocellular carcinoma (HCC) by activation of hepatic stellate cells and modulation of hepatocyte differentiation. Shedding of Axl’s ectodomain leads to the release of soluble Axl (sAxl), which is increased in advanced fibrosis and in early-to-late stage HCC in the presence and absence of cirrhosis. Here, we focus on the dynamics of Axl receptor shedding and delineate possible scenarios how Axl signaling might act as driver of fibrosis progression and HCC development. Based on experimental and clinical data, we discuss the consequences of modifying Axl signaling by sAxl cleavage, as well as cellular strategies to escape from antagonizing effects of Axl shedding by the involvement of the hepatic microenvironment. We emphasize a correlation between free Gas6 and free sAxl levels favoring abundant Gas6/Axl signaling in advanced fibrosis and HCC. The raised scenario provides a solid basis for theranostics allowing the use of sAxl as an accurate diagnostic biomarker of liver cirrhosis and HCC, as well as Axl receptor signaling for therapeutic intervention in stratified HCC patients.
Collapse
|
30
|
Nahar S, Nakashima Y, Miyagi-Shiohira C, Kinjo T, Toyoda Z, Kobayashi N, Saitoh I, Watanabe M, Noguchi H, Fujita J. Cytokines in adipose-derived mesenchymal stem cells promote the healing of liver disease. World J Stem Cells 2018; 10:146-159. [PMID: 30631390 PMCID: PMC6325075 DOI: 10.4252/wjsc.v10.i11.146] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/07/2018] [Accepted: 10/11/2018] [Indexed: 02/06/2023] Open
Abstract
Adipose-derived mesenchymal stem cells (ADSCs) are a treatment cell source for patients with chronic liver injury. ADSCs are characterized by being harvested from the patient's own subcutaneous adipose tissue, a high cell yield (i.e., reduced immune rejection response), accumulation at a disease nidus, suppression of excessive immune response, production of various growth factors and cytokines, angiogenic effects, anti-apoptotic effects, and control of immune cells via cell-cell interaction. We previously showed that conditioned medium of ADSCs promoted hepatocyte proliferation and improved the liver function in a mouse model of acute liver failure. Furthermore, as found by many other groups, the administration of ADSCs improved liver tissue fibrosis in a mouse model of liver cirrhosis. A comprehensive protein expression analysis by liquid chromatography with tandem mass spectrometry showed that the various cytokines and chemokines produced by ADSCs promote the healing of liver disease. In this review, we examine the ability of expressed protein components of ADSCs to promote healing in cell therapy for liver disease. Previous studies demonstrated that ADSCs are a treatment cell source for patients with chronic liver injury. This review describes the various cytokines and chemokines produced by ADSCs that promote the healing of liver disease.
Collapse
Affiliation(s)
- Saifun Nahar
- Department of Infectious, Respiratory, and Digestive Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Yoshiki Nakashima
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Chika Miyagi-Shiohira
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Takao Kinjo
- Department of Basic Laboratory Sciences, School of Health Sciences in the Faculty of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Zensei Toyoda
- Department of Basic Laboratory Sciences, School of Health Sciences in the Faculty of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | | | - Issei Saitoh
- Division of Pediatric Dentistry, Graduate School of Medical and Dental Science, Niigata University, Niigata 951-8514, Japan
| | - Masami Watanabe
- Department of Urology, Okayama Univer sity Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hirofumi Noguchi
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | - Jiro Fujita
- Department of Infectious, Respiratory, and Digestive Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| |
Collapse
|
31
|
Growth arrest-specific gene 6 transfer promotes mesenchymal stem cell survival and cardiac repair under hypoxia and ischemia via enhanced autocrine signaling and paracrine action. Arch Biochem Biophys 2018; 660:108-120. [PMID: 30365934 DOI: 10.1016/j.abb.2018.10.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 10/08/2018] [Accepted: 10/22/2018] [Indexed: 01/10/2023]
Abstract
Poor cell viability after transplantation has restricted the therapeutic capacity of mesenchymal stem cells (MSCs) for cardiac dysfunction after myocardial infarction (MI). Growth arrest-specific gene 6 (Gas6) encodes a secreted γ-carboxyglutamic acid (Gla)-containing protein that functions in cell growth, adhesion, chemotaxis, mitogenesis and cell survival. In this study, we genetically modified MSCs with Gas6 and evaluated cell survival, cardiac function, and infarct size in a rat model of MI via intramyocardial delivery. Functional studies demonstrated that Gas6 transfer significantly reduced MSC apoptosis, increased survival of MSCs in vitro and in vivo, and that Gas6-engineered MSCs (MSCGas6)-treated animals had smaller infarct size and showed remarkably functional recovery as compared with control MSCs (MSCNull)-treated animals. Mechanistically, Gas6 could enhance phosphatidylinositol 3-kinase (PI3K)/Akt signaling and improve hypoxia-inducible factor-1 alpha (HIF-1α)-driven secretion of four major growth factors (VEGF, bFGF, SDF and IGF-1) in MSCs under hypoxia in an Axl-dependent autocrine manner. The paracrine action of MSCGas6 was further validated by coculture neonatal rat cardiomyocytes with conditioned medium from hypoxia-treated MSCGas6, as well as by pretreatment cardiomyocytes with the specific receptor inhibitors of VEGF, bFGF, SDF and IGF-1. Collectively, our data suggest that Gas6 may advance the efficacy of MSC therapy for post-infarcted heart failure via enhanced Gas6/Axl autocrine prosurvival signaling and paracrine cytoprotective action.
Collapse
|
32
|
Bellan M, Castello LM, Pirisi M. Candidate Biomarkers of Liver Fibrosis: A Concise, Pathophysiology-oriented Review. J Clin Transl Hepatol 2018; 6:317-325. [PMID: 30271745 PMCID: PMC6160308 DOI: 10.14218/jcth.2018.00006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/25/2018] [Accepted: 05/03/2018] [Indexed: 12/11/2022] Open
Abstract
Repair of sustained liver injury results in fibrosis (i.e. the accumulation of extracellular matrix proteins), and ultimately the complete distortion of parenchymal architecture of the liver, which we call cirrhosis. Detecting and staging of fibrosis is thus a mainstay in the management of chronic liver diseases, since many clinically relevant decisions, such as starting treatment and/or monitoring for complications including hepatocellular carcinoma, may depend on it. The gold standard for fibrosis staging is liver biopsy, the role of which, however, is questioned nowadays because of cost, hazards and poor acceptance by patients. On the other hand, imaging techniques and/or measurement of direct and indirect serum markers have not proved to be completely satisfactory under all circumstances as alternatives to liver biopsy. Making progress in this field is now more crucial than ever, since treatments for established fibrosis appear on the horizon. Fine dissection of the pathways involved in the pathophysiology of liver diseases has put forward several novel candidate biomarkers of liver fibrosis, such as growth arrest-specific6, Mac-2-binding protein, osteopontin, placental growth factor, growth/differentiation factor 15 and hepatocyte growth factor. All molecules have been suggested to have potential to complement or substitute methods currently used to stage liver diseases. Here, we review the pros and cons for their use in this setting.
Collapse
Affiliation(s)
- Mattia Bellan
- Department of Translational Medicine, Università del Piemonte Orientale UPO, Novara, Italy
- Division of Internal Medicine, “Sant’Andrea Hospital”, Vercelli, Italy
- IRCAD, Interdisciplinary Research Center of Autoimmune Diseases, Novara, Italy
- *Correspondence to: Mattia Bellan, Department of Translational Medicine, Università del Piemonte Orientale UPO, via Solaroli 17, Novara (NO) 28100, Italy. Tel: +39-321-3733966, Fax: +39-321-3733361, E-mail:
| | - Luigi Mario Castello
- Department of Translational Medicine, Università del Piemonte Orientale UPO, Novara, Italy
- Emergency Medicine Department, “AOU Maggiore della Carità”, Novara, Italy
| | - Mario Pirisi
- Department of Translational Medicine, Università del Piemonte Orientale UPO, Novara, Italy
- Division of Internal Medicine, “AOU Maggiore della Carità, Novara, Italy
| |
Collapse
|
33
|
Zhang G, Wang M, Zhao H, Cui W. Function of Axl receptor tyrosine kinase in non-small cell lung cancer. Oncol Lett 2017; 15:2726-2734. [PMID: 29434997 DOI: 10.3892/ol.2017.7694] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 11/07/2017] [Indexed: 11/06/2022] Open
Abstract
Axl receptor tyrosine kinase (hereafter Axl) is a member of the tyrosine-protein kinase receptor Tyro3, Axl and proto-oncogene tyrosine-protein kinase Mer family of receptor tyrosine kinases, possessing multiple different functions in normal cells. Axl is overexpressed and activated in numerous different human cancer types, triggering several signaling pathways and enhancing tumor progression. The present review assesses previous studies on the function of Axl in non-small cell lung cancer (NSCLC). Axl is overexpressed in the tumor tissues of a number of patients with NSCLC and is associated with poorer clinical outcomes; it promotes NSCLC tumor growth, invasion/metastasis, drug resistance and the epithelial-mesenchymal transition, thus providing a survival advantage to tumor cells. Therefore, Axl may be a promising target in NSCLC treatment.
Collapse
Affiliation(s)
- Guoan Zhang
- Cancer Pathology Research Institute, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Meng Wang
- Department of Oncology, Shandong Jining First People's Hospital, Jining, Shandong 272111, P.R. China
| | - Hongli Zhao
- Department of Gastroenterology, Shandong Control Center for Digestive Diseases, Jining, Shandong 272033, P.R. China
| | - Wen Cui
- Cancer Pathology Research Institute, Jining Medical University, Jining, Shandong 272067, P.R. China
| |
Collapse
|
34
|
Dengler M, Staufer K, Huber H, Stauber R, Bantel H, Weiss KH, Starlinger P, Pock H, Klöters-Plachky P, Gotthardt DN, Rauch P, Lackner C, Stift J, Brostjan C, Gruenberger T, Kumada T, Toyoda H, Tada T, Weiss TS, Trauner M, Mikulits W. Soluble Axl is an accurate biomarker of cirrhosis and hepatocellular carcinoma development: results from a large scale multicenter analysis. Oncotarget 2017; 8:46234-46248. [PMID: 28526812 PMCID: PMC5542263 DOI: 10.18632/oncotarget.17598] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 04/06/2017] [Indexed: 12/21/2022] Open
Abstract
Patients with chronic liver disease (CLD) and cirrhosis are at high risk for hepatocellular carcinoma (HCC). Current diagnostic tools for HCC detection include imaging techniques and serum biomarkers such as α-fetoprotein (AFP). Yet, these methods are limited in sensitivity and specificity to accurately detect early HCC. Here we focused on the potential of soluble Axl (sAxl) as a biomarker in CLD patients by analyzing serum samples of 1067 patients and healthy controls from centers in Europe and Asia. We show that serum concentrations of sAxl were significantly increased at early (82.57 ng/mL) and later stages of HCC (114.50 ng/mL) as compared to healthy controls (40.15 ng/mL). Notably, no elevated sAxl levels were detected in patients with CLD including chronic viral hepatitis, autoimmune hepatitis, cholestatic liver disease, or non-alcoholic fatty liver disease versus healthy controls. Furthermore, sAxl did not rise in liver adenomas or cholangiocarcinoma (CCA). Yet, patients with advanced fibrosis (F3) or cirrhosis (F4) showed enhanced sAxl concentrations (F3: 54.67 ng/mL; F4: 94.74 ng/mL). Hepatic myofibroblasts exhibited an increased release of sAxl, suggesting that elevated sAxl levels arise from these cells during fibrosis. Receiver operating characteristic curve analysis of sAxl displayed a strongly increased sensitivity and specificity to detect both cirrhosis (80.8%/92.0%) and HCC (83.3%/86.7%) with an area under the curve of 0.935/0.903 as compared to AFP. In conclusion, sAxl shows high diagnostic accuracy at early stage HCC as well as cirrhosis, thereby outperforming AFP. Importantly, sAxl remains normal in most common CLDs, liver adenomas and CCA.
Collapse
Affiliation(s)
- Mirko Dengler
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Katharina Staufer
- Department of Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Heidemarie Huber
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Rudolf Stauber
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Heike Bantel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | | | - Patrick Starlinger
- Department of Surgery, Division of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Hannelore Pock
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | | | | - Peter Rauch
- Candor Bioscience GmbH, Wangen im Allgäu, Germany
| | - Carolin Lackner
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Judith Stift
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Christine Brostjan
- Department of Surgery, Division of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Thomas Gruenberger
- Department of Surgery, Division of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Takashi Kumada
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Toshifumi Tada
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Thomas S. Weiss
- Center for Liver Cell Research, Children's University Hospital (KUNO), University of Regensburg Hospital, Regensburg, Germany
| | - Michael Trauner
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Mikulits
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
35
|
Abstract
Hepatic fibrosis is a dynamic process characterized by the net accumulation of extracellular matrix resulting from chronic liver injury of any aetiology, including viral infection, alcoholic liver disease and NASH. Activation of hepatic stellate cells (HSCs) - transdifferentiation of quiescent, vitamin-A-storing cells into proliferative, fibrogenic myofibroblasts - is now well established as a central driver of fibrosis in experimental and human liver injury. Yet, the continued discovery of novel pathways and mediators, including autophagy, endoplasmic reticulum stress, oxidative stress, retinol and cholesterol metabolism, epigenetics and receptor-mediated signals, reveals the complexity of HSC activation. Extracellular signals from resident and inflammatory cells including macrophages, hepatocytes, liver sinusoidal endothelial cells, natural killer cells, natural killer T cells, platelets and B cells further modulate HSC activation. Finally, pathways of HSC clearance have been greatly clarified, and include apoptosis, senescence and reversion to an inactivated state. Collectively, these findings reinforce the remarkable complexity and plasticity of HSC activation, and underscore the value of clarifying its regulation in hopes of advancing the development of novel diagnostics and therapies for liver disease.
Collapse
Affiliation(s)
- Takuma Tsuchida
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1123, New York, New York 10029, USA.,Research Division, Mitsubishi Tanabe Pharma Corporation, 2-2-50, Kawagishi, Toda-shi, Saitama 335-8505, Japan
| | - Scott L Friedman
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1123, New York, New York 10029, USA
| |
Collapse
|
36
|
Dihingia A, Kalita J, Manna P. Implication of a novel Gla-containing protein, Gas6 in the pathogenesis of insulin resistance, impaired glucose homeostasis, and inflammation: A review. Diabetes Res Clin Pract 2017; 128:74-82. [PMID: 28453960 DOI: 10.1016/j.diabres.2017.03.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 03/28/2017] [Indexed: 12/15/2022]
Abstract
Growth arrest specific 6 (Gas6), a vitamin K-dependent protein plays a significant role in the regulation of cellular homeostasis via binding with TAM-receptor tyrosine kinases. Several studies reported the role of Gas6 in cancer, glomerular injury, obesity, and inflammation, however, very little is known about its role in insulin resistance (IR) and impaired glucose metabolism. Majority of the studies reported an inverse correlation of Gas6 protein levels or gene polymorphism with plasma glucose, HbA1c, IR, and inflammatory cytokines among type 2 diabetes (T2D) and obese subjects. However, few studies reported a positive correlation of Gas6 protein levels or gene polymorphism with IR and inflammation among obese subjects. This review for the first time provides an overview of the association of Gas6 protein levels or gene polymorphism with IR, glucose intolerance, and inflammation among T2D and obese subjects. This review also depicts the probable mechanism underlying the association of Gas6 with glucose intolerance and inflammation. The outcome of this review will increase the understanding about the role of Gas6 in the pathogenesis of IR, glucose intolerance and inflammation and that should in turn lead to the design of clinical interventions to improve glucose metabolism and the lives of the T2D patients.
Collapse
Affiliation(s)
- Anjum Dihingia
- Academy of Scientific and Innovative Research, Chennai 600113, India; Biological Science and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat, Assam 785006, India
| | - Jatin Kalita
- Academy of Scientific and Innovative Research, Chennai 600113, India; Biological Science and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat, Assam 785006, India
| | - Prasenjit Manna
- Academy of Scientific and Innovative Research, Chennai 600113, India; Biological Science and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat, Assam 785006, India.
| |
Collapse
|
37
|
Uehara S, Fukuzawa Y, Matuyama T, Gotoh K. Role of Tyro3, Axl, and Mer Receptors and Their Ligands (Gas6, and Protein S) in Patients with Hepatocellular Carcinoma. ACTA ACUST UNITED AC 2017. [DOI: 10.4236/jct.2017.82010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
38
|
Bellan M, Pogliani G, Marconi C, Minisini R, Franzosi L, Alciato F, Magri A, Avanzi GC, Pirisi M, Sainaghi PP. Gas6 as a putative noninvasive biomarker of hepatic fibrosis. Biomark Med 2016; 10:1241-1249. [PMID: 27924629 DOI: 10.2217/bmm-2016-0210] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
AIM To evaluate serum growth arrest-specific gene 6 (Gas6) concentration as a biomarker of liver fibrosis progression. MATERIALS & METHODS One hundred and thirteen consecutive patients affected by chronic liver disease underwent transient elastography, Gas6 measurement and, if clinically indicated, liver biopsy. RESULTS Gas6 concentration was directly correlated to liver stiffness (r = 0.67; p < 0.0001) and was significantly higher in patients with advanced fibrosis (Ishak 4-5; p < 0.001). A plasma concentration <30 ng/ml Gas6 ruled out fibrosis with 84% sensitivity and 56% specificity, while values >42 ng/ml identified severe fibrosis with a sensitivity of 64% and a specificity of 95%; the diagnostic accuracy was comparable to that of transient elastography. CONCLUSION Gas6 is a novel biomarker of liver fibrosis, with a potential clinical and pathophysiological relevance.
Collapse
Affiliation(s)
- Mattia Bellan
- Department of Translational Medicine, Università del Piemonte Orientale UPO, via Solaroli 17, 28100 Novara, Italy.,Division of Internal Medicine, "AOU Maggiore della Carità", Corso Mazzini 18, 28100 Novara, Italy
| | - Gabriele Pogliani
- Department of Translational Medicine, Università del Piemonte Orientale UPO, via Solaroli 17, 28100 Novara, Italy.,Division of Internal Medicine, "AOU Maggiore della Carità", Corso Mazzini 18, 28100 Novara, Italy
| | - Cecilia Marconi
- Department of Translational Medicine, Università del Piemonte Orientale UPO, via Solaroli 17, 28100 Novara, Italy.,Division of Internal Medicine, "AOU Maggiore della Carità", Corso Mazzini 18, 28100 Novara, Italy
| | - Rosalba Minisini
- Department of Translational Medicine, Università del Piemonte Orientale UPO, via Solaroli 17, 28100 Novara, Italy.,Division of Internal Medicine, "AOU Maggiore della Carità", Corso Mazzini 18, 28100 Novara, Italy
| | - Lisa Franzosi
- Department of Translational Medicine, Università del Piemonte Orientale UPO, via Solaroli 17, 28100 Novara, Italy.,Division of Internal Medicine, "AOU Maggiore della Carità", Corso Mazzini 18, 28100 Novara, Italy
| | - Federica Alciato
- Department of Translational Medicine, Università del Piemonte Orientale UPO, via Solaroli 17, 28100 Novara, Italy
| | - Andrea Magri
- Department of Translational Medicine, Università del Piemonte Orientale UPO, via Solaroli 17, 28100 Novara, Italy.,Division of Internal Medicine, "AOU Maggiore della Carità", Corso Mazzini 18, 28100 Novara, Italy
| | - Gian Carlo Avanzi
- Department of Translational Medicine, Università del Piemonte Orientale UPO, via Solaroli 17, 28100 Novara, Italy.,Emergency Medicine Department, "AOU Maggiore della Carità", Corso Mazzini 18, Novara, Italy
| | - Mario Pirisi
- Department of Translational Medicine, Università del Piemonte Orientale UPO, via Solaroli 17, 28100 Novara, Italy.,Division of Internal Medicine, "AOU Maggiore della Carità", Corso Mazzini 18, 28100 Novara, Italy.,IRCAD, Interdisciplinary Research Center of Autoimmune Diseases, via Solaroli 17, Novara, Italy
| | - Pier Paolo Sainaghi
- Division of Internal Medicine, "AOU Maggiore della Carità", Corso Mazzini 18, 28100 Novara, Italy.,IRCAD, Interdisciplinary Research Center of Autoimmune Diseases, via Solaroli 17, Novara, Italy
| |
Collapse
|
39
|
Davra V, Kimani SG, Calianese D, Birge RB. Ligand Activation of TAM Family Receptors-Implications for Tumor Biology and Therapeutic Response. Cancers (Basel) 2016; 8:cancers8120107. [PMID: 27916840 PMCID: PMC5187505 DOI: 10.3390/cancers8120107] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 11/16/2016] [Accepted: 11/26/2016] [Indexed: 12/21/2022] Open
Abstract
The TAM family of receptors (i.e., Tyro3, Axl, and Mertk), and their ligands Growth arrest specific factor 6 (Gas6) and Protein S (Pros1) contribute to several oncogenic processes, such as cell survival, invasion, migration, chemo-resistance, and metastasis, whereby expression often correlates with poor clinical outcomes. In recent years, there has been great interest in the study of TAM receptors in cancer, stemming both from their roles as oncogenic signaling receptors, as well as their roles in tumor immunology. As a result, several classes of TAM inhibitors that include small molecule tyrosine kinase inhibitors, monoclonal antibodies, decoy receptors, as well as novel strategies to target TAM ligands are being developed. This paper will review the biology of TAM receptors and their ligands with a focus on cancer, as well as evidence-based data for the continued pursuit of TAM/Gas6 inhibitors in clinical practice.
Collapse
Affiliation(s)
- Viralkumar Davra
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ 07103, USA.
| | - Stanley G Kimani
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ 07103, USA.
| | - David Calianese
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ 07103, USA.
| | - Raymond B Birge
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ 07103, USA.
| |
Collapse
|
40
|
The Gas6/TAM System and Multiple Sclerosis. Int J Mol Sci 2016; 17:ijms17111807. [PMID: 27801848 PMCID: PMC5133808 DOI: 10.3390/ijms17111807] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/22/2016] [Accepted: 10/26/2016] [Indexed: 01/25/2023] Open
Abstract
Growth arrest specific 6 (Gas6) is a multimodular circulating protein, the biological actions of which are mediated by the interaction with three transmembrane tyrosine kinase receptors: Tyro3, Axl, and MerTK, collectively named TAM. Over the last few decades, many progresses have been done in the understanding of the biological activities of this highly pleiotropic system, which plays a role in the regulation of immune response, inflammation, coagulation, cell growth, and clearance of apoptotic bodies. Recent findings have further related Gas6 and TAM receptors to neuroinflammation in general and, specifically, to multiple sclerosis (MS). In this paper, we review the biology of the Gas6/TAM system and the current evidence supporting its potential role in the pathogenesis of MS.
Collapse
|
41
|
Petta S, Marra F. Reply to 'Genetic and clinical data reinforce the role of GAS6 and TAM receptors in liver fibrosis'. J Hepatol 2016; 64:984-5. [PMID: 26812076 DOI: 10.1016/j.jhep.2016.01.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 01/21/2016] [Indexed: 01/26/2023]
Affiliation(s)
- Salvatore Petta
- Sezione di Gastroenterologia, DiBiMIS, University of Palermo, Italy.
| | - Fabio Marra
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Italy
| |
Collapse
|
42
|
Petta S, Valenti L, Marra F, Grimaudo S, Tripodo C, Bugianesi E, Cammà C, Cappon A, Di Marco V, Di Maira G, Dongiovanni P, Rametta R, Gulino A, Mozzi E, Orlando E, Maggioni M, Pipitone RM, Fargion S, Craxì A. MERTK rs4374383 polymorphism affects the severity of fibrosis in non-alcoholic fatty liver disease. J Hepatol 2016; 64:682-90. [PMID: 26596542 DOI: 10.1016/j.jhep.2015.10.016] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 09/16/2015] [Accepted: 10/12/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIM Homozygosity for a common non-coding rs4374383 G>A polymorphism in MERTK (myeloid-epithelial-reproductive tyrosine kinase) has been associated with the protection against fibrosis progression in chronic hepatitis C. The main study objective was to assess whether MERTK AA genotype influences liver fibrosis, and secondarily MERTK expression in patients with non-alcoholic fatty liver disease (NAFLD). We also investigated whether MERTK is expressed in human hepatic stellate cells (HSC) and in murine models of fibrogenesis. METHODS We considered 533 consecutive patients who underwent liver biopsy for suspected non-alcoholic steatohepatitis (NASH) without severe obesity from two Italian cohorts. As controls, we evaluated 158 patients with normal liver enzymes and without metabolic disturbances. MERTK rs4374383 genotype was assessed by 5'-nuclease assays. MERTK expression was analysed in mouse models of fibrosis, and the effect of the MERTK ligand GAS6 were investigated in human HSC. RESULTS Clinically significant fibrosis (stage F2-F4) was observed in 19% of patients with MERTK AA compared to 30% in those with MERTK GG/GA (OR 0.43, CI 0.21-0.88, p=0.02; adjusted for centre, and genetic, clinical-metabolic and histological variables). The protective rs4374383 AA genotype was associated with lower MERTK hepatic expression. MERTK was overexpressed in the liver of NAFLD patients with F2-F4 fibrosis and in in vivo models of fibrogenesis. Furthermore, exposure of cultured human HSC to the MERTK ligand GAS6, increased cell migration and induced procollagen expression. These effects were counteracted by inhibition of MERTK activity, which also resulted in apoptotic death of HSC. CONCLUSIONS The rs4374383 AA genotype, associated with lower intrahepatic expression of MERTK, is protective against F2-F4 fibrosis in patients with NAFLD. The mechanism may involve modulation of HSC activation.
Collapse
Affiliation(s)
- Salvatore Petta
- Sezione di Gastroenterologia, DiBiMIS, University of Palermo, Italy.
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi, Internal Medicine, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - Fabio Marra
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Italy
| | | | - Claudio Tripodo
- Cattedra di Anatomia Patologica, University of Palermo, Italy
| | - Elisabetta Bugianesi
- Division of Gastro-Hepatology, Department of Medical Sciences, San Giovanni Battista Hospital, University of Torino, Torino, Italy
| | - Calogero Cammà
- Sezione di Gastroenterologia, DiBiMIS, University of Palermo, Italy
| | - Andrea Cappon
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Italy
| | - Vito Di Marco
- Sezione di Gastroenterologia, DiBiMIS, University of Palermo, Italy
| | - Giovanni Di Maira
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Italy
| | - Paola Dongiovanni
- Department of Pathophysiology and Transplantation, Università degli Studi, Internal Medicine, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - Raffaela Rametta
- Department of Pathophysiology and Transplantation, Università degli Studi, Internal Medicine, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | | | - Enrico Mozzi
- Department of Pathophysiology and Transplantation, Università degli Studi, Internal Medicine, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - Emanuele Orlando
- Sezione di Gastroenterologia, DiBiMIS, University of Palermo, Italy
| | - Marco Maggioni
- Pathology, Fondazione IRCCS Ca' Granda Policlinico, Milano, Italy
| | | | - Silvia Fargion
- Department of Pathophysiology and Transplantation, Università degli Studi, Internal Medicine, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - Antonio Craxì
- Sezione di Gastroenterologia, DiBiMIS, University of Palermo, Italy
| |
Collapse
|
43
|
Bárcena C, Stefanovic M, Tutusaus A, Joannas L, Menéndez A, García-Ruiz C, Sancho-Bru P, Marí M, Caballeria J, Rothlin CV, Fernández-Checa JC, de Frutos PG, Morales A. Gas6/Axl pathway is activated in chronic liver disease and its targeting reduces fibrosis via hepatic stellate cell inactivation. J Hepatol 2015; 63:670-8. [PMID: 25908269 PMCID: PMC4543529 DOI: 10.1016/j.jhep.2015.04.013] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 04/07/2015] [Accepted: 04/10/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Liver fibrosis, an important health concern associated to chronic liver injury that provides a permissive environment for cancer development, is characterized by accumulation of extracellular matrix components mainly derived from activated hepatic stellate cells (HSCs). Axl, a receptor tyrosine kinase and its ligand Gas6, are involved in cell differentiation, immune response and carcinogenesis. METHODS HSCs were obtained from WT and Axl(-/-) mice, treated with recombinant Gas6 protein (rGas6), Axl siRNAs or the Axl inhibitor BGB324, and analyzed by western blot and real-time PCR. Experimental fibrosis was studied in CCl4-treated WT and Axl(-/-) mice, and in combination with Axl inhibitor. Gas6 and Axl serum levels were measured in alcoholic liver disease (ALD) and hepatitis C virus (HCV) patients. RESULTS In primary mouse HSCs, Gas6 and Axl levels paralleled HSC activation. rGas6 phosphorylated Axl and AKT prior to HSC phenotypic changes, while Axl siRNA silencing reduced HSC activation. Moreover, BGB324 blocked Axl/AKT phosphorylation and diminished HSC activation. In addition, Axl(-/-) mice displayed decreased HSC activation in vitro and liver fibrogenesis after chronic damage by CCl4 administration. Similarly, BGB324 reduced collagen deposition and CCl4-induced liver fibrosis in mice. Importantly, Gas6 and Axl serum levels increased in ALD and HCV patients, inversely correlating with liver functionality. CONCLUSIONS The Gas6/Axl axis is required for full HSC activation. Gas6 and Axl serum levels increase in parallel to chronic liver disease progression. Axl targeting may be a therapeutic strategy for liver fibrosis management.
Collapse
Affiliation(s)
- Cristina Bárcena
- Liver Unit, Hospital Clinic, IDIBAPS-CIBEK, CIBEREHD, Barcelona, Catalonia, Sain,Department of Cell Death and Proliferation, IIBB-CSIC, Barcelona, Catalonia, Spain
| | - Milica Stefanovic
- Liver Unit, Hospital Clinic, IDIBAPS-CIBEK, CIBEREHD, Barcelona, Catalonia, Sain,Department of Cell Death and Proliferation, IIBB-CSIC, Barcelona, Catalonia, Spain
| | - Anna Tutusaus
- Liver Unit, Hospital Clinic, IDIBAPS-CIBEK, CIBEREHD, Barcelona, Catalonia, Sain,Department of Cell Death and Proliferation, IIBB-CSIC, Barcelona, Catalonia, Spain
| | - Leonel Joannas
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Anghara Menéndez
- Department of Cell Death and Proliferation, IIBB-CSIC, Barcelona, Catalonia, Spain
| | - Carmen García-Ruiz
- Liver Unit, Hospital Clinic, IDIBAPS-CIBEK, CIBEREHD, Barcelona, Catalonia, Sain,Department of Cell Death and Proliferation, IIBB-CSIC, Barcelona, Catalonia, Spain
| | - Pau Sancho-Bru
- Liver Unit, Hospital Clinic, IDIBAPS-CIBEK, CIBEREHD, Barcelona, Catalonia, Sain
| | - Montserrat Marí
- Liver Unit, Hospital Clinic, IDIBAPS-CIBEK, CIBEREHD, Barcelona, Catalonia, Sain,Department of Cell Death and Proliferation, IIBB-CSIC, Barcelona, Catalonia, Spain
| | - Joan Caballeria
- Liver Unit, Hospital Clinic, IDIBAPS-CIBEK, CIBEREHD, Barcelona, Catalonia, Sain
| | - Carla V. Rothlin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - José C. Fernández-Checa
- Liver Unit, Hospital Clinic, IDIBAPS-CIBEK, CIBEREHD, Barcelona, Catalonia, Sain,Department of Cell Death and Proliferation, IIBB-CSIC, Barcelona, Catalonia, Spain,Research Center for Alcoholic Liver and Pancreatic Diseases, Keck School of Medicine of the University of Southern California, Los Angeles, CA USA
| | | | - Albert Morales
- Liver Unit, Hospital Clinic, IDIBAPS-CIBEK, CIBEREHD, Barcelona, Catalonia, Spain; Department of Cell Death and Proliferation, IIBB-CSIC, Barcelona, Catalonia, Spain.
| |
Collapse
|
44
|
SDF-1/CXCR4 Axis Promotes MSCs to Repair Liver Injury Partially through Trans-Differentiation and Fusion with Hepatocytes. Stem Cells Int 2015; 2015:960387. [PMID: 26300925 PMCID: PMC4537768 DOI: 10.1155/2015/960387] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 03/06/2015] [Indexed: 02/06/2023] Open
Abstract
MSCs have become a popular target for developing end-stage liver therapies. In this study, two models of bone marrow chimeric mice were used to construct the liver failure models. Then it was found that MSCs can transdifferentiate into hepatocyte-like cells and these hepatocyte-like cells can significantly express albumin. Furthermore it was also found that MSCs can fuse with the hepatocytes and these cells had the proliferation activity. However, the percentage of transdifferentiation was significantly higher than fusion. So it was considered that MSCs which transdifferentiated into hepatocyte-likes cells played important roles for repairing the injuring liver function.
Collapse
|
45
|
Chiu KC, Lee CH, Liu SY, Yeh CT, Huang RY, Yuh DY, Cheng JC, Chou YT, Shieh YS. Protumoral effect of macrophage through Axl activation on mucoepidermoid carcinoma. J Oral Pathol Med 2015; 43:538-44. [PMID: 25184164 DOI: 10.1111/jop.12163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study aims to test the potential involvement of Axl signaling in the protumoral effect of tumor-associated macrophages (TAMs) in mucoepidermoid carcinoma (MEC). MATERIALS AND METHODS We carried out cocultured experiments by incubation of MEC cells (UTMUC-1) and macrophages (THP-1) and examined Axl activation status. The expression of MMPs and behavior change were examined in UT-MUC-1 cells. The effect of Axl signaling on co-cultured cancer cells was further investigated by knockdown Axl expression and suppression by Axl-specific inhibitor R428. RESULTS Activation of Axl signaling and increased expression and activity of MMP-2 and MMP-9 along with increased invasion/migration ability in MEC cells were observed when co-cultured with TAMs. Upon knockdown of Axl in MEC or addition of R428 in the co-cultured system, these co-cultured effects were diminished. CONCLUSION TAMs play a protumoral role in MEC via activation of the Axl signaling pathway, up-regulating MMPs expression, and increasing invasion/migration ability.
Collapse
|
46
|
Abstract
The control of cellular growth and proliferation is key to the maintenance of homeostasis. Survival, proliferation, and arrest are regulated, in part, by Growth Arrest Specific 6 (Gas6) through binding to members of the TAM receptor tyrosine kinase family. Activation of the TAM receptors leads to downstream signaling through common kinases, but the exact mechanism within each cellular context varies and remains to be completely elucidated. Deregulation of the TAM family, due to its central role in mediating cellular proliferation, has been implicated in multiple diseases. Axl was cloned as the first TAM receptor in a search for genes involved in the progression of chronic to acute-phase leukemia, and has since been established as playing a critical role in the progression of cancer. The oncogenic nature of Axl is demonstrated through its activation of signaling pathways involved in proliferation, migration, inhibition of apoptosis, and therapeutic resistance. Despite its recent discovery, significant progress has been made in the development of effective clinical therapeutics targeting Axl. In order to accurately define the role of Axl in normal and diseased processes, it must be analyzed in a cell type-specific context.
Collapse
|
47
|
Qi F, Hu JF, Tang D, Liu BH, Zhu L. Identification of differential gene expression profile of HSCs subjected to pressurization by next-generation sequencing. Shijie Huaren Xiaohua Zazhi 2014; 22:3380-3387. [DOI: 10.11569/wcjd.v22.i23.3380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To identify the gene expression profile of hepatic stellate cells (HSCs) subjected to pressurization by next-generation sequencing.
METHODS: Primary HSCs were isolated and cultured. After cultivation for 14 d, cells were subjected to a pressure of 10 MMHG (millimeters of mercury) for 1 h. Total RNA was then extracted and reverse transcribed into cDNA to screen differential genes between HSCs subjected to pressurization and non-treated HSCs using DEG-Seq.
RESULTS: A total of 979 differentially expressed genes were expressed in HSCs, of which 14 showed the most significant difference, including 10 up-regulated and 4 down-regulated genes. These genes were found to be related to anabolism, immune response, cell apoptosis, etc.
CONCLUSION: Differentially expressed genes could be identified in HSCs subjected to pressurization, indicating that HSCs respond to external pressure stimulation by regulating the expression of these genes.
Collapse
|
48
|
Lee CH, Liu SY, Chou KC, Yeh CT, Shiah SG, Huang RY, Cheng JC, Yen CY, Shieh YS. Tumor-associated macrophages promote oral cancer progression through activation of the Axl signaling pathway. Ann Surg Oncol 2014; 21:1031-7. [PMID: 24276640 DOI: 10.1245/s10434-013-3400-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND Recent studies suggest that tumor-associated macrophages (TAMs) promote tumor growth and metastasis. Our previous report demonstrated that Axl signaling promotes carcinogenesis and progression of oral squamous cell carcinoma (OSCC). This study aims to test the potential involvement of growth arrest-specific gene 6 (Gas6)/Axl signaling in the protumoral effect of TAMs. METHODS Co-culture experiments by incubation of OSCC cells (YD38 and OE) and macrophages (THP-1) were performed. The expression of Gas6/Axl and epithelial-mesenchymal transition (EMT) genes were examined in YD38 and OE cells. The effect of Gas6/Axl signaling on co-cultured cancer cells was further investigated by knocking down Axl expression and neutralizing Gas6. Axl and TAM distribution were analyzed by immunohistochemistry in OSCC tissues. RESULTS Activation of Axl signaling and increased expression of mesenchymal markers, along with increased invasion/migration ability of OSCC cells, was noted upon co-culture with THP-1. Neutralization of Gas6 in the co-culture system or knockdown of Axl in YD38 caused the co-culture effects to be diminished. Co-culture with THP-1 increased nuclear factor (NF)-κB nuclear translocation and transcription activity in YD38 cells. A significant association between the TAM count and expression of phosphorylated Axl (P = 0.004) was found in vivo cancer tissues. CONCLUSIONS TAMs play a protumor role in OSCC and likely promote tumor progression through activation of the Gas6/Axl-NF-κB signaling pathway. Therefore, Gas6/Axl and NF-κB signaling in OSCC cells may be a putative target for therapeutic intervention.
Collapse
Affiliation(s)
- Chien-Hsing Lee
- Department of Internal Medicine,Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Haase TN, Rasmussen M, Jaksch CAM, Gaarn LW, Petersen CK, Billestrup N, Nielsen JH. Growth arrest specific protein (GAS) 6: a role in the regulation of proliferation and functional capacity of the perinatal rat beta cell. Diabetologia 2013; 56:763-73. [PMID: 23334461 DOI: 10.1007/s00125-012-2821-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 12/13/2012] [Indexed: 11/28/2022]
Abstract
AIMS/HYPOTHESIS Maternal low-protein (LP) diet during gestation results in a reduced beta cell mass in the offspring at birth and this may hamper the ability to adapt to high-energy food and sedentary lifestyle later in life. To investigate the biology behind the LP-offspring phenotype, this study aimed to identify differentially expressed genes in the pancreas and their potential role in the fetal programming. METHODS Wistar rats were given either an LP diet or normal-chow (NC) diet during gestation and differentially expressed genes in the offspring around the time of birth were identified using RNA microarray and quantitative PCR. The role of a differentially expressed gene, growth arrest specific protein 6 (GAS6), was evaluated in vitro using neonatal rat islets. RESULTS The mRNA level of Gas6, known to be mitogenic in other tissues, was reduced in LP offspring. The mRNA content of Mafa was increased in LP offspring suggesting an early maturation of beta cells. When applied in vitro, GAS6 increased proliferation of neonatal pancreatic beta cells, while reducing glucose-stimulated insulin secretion without changing the total insulin content of the islets. In addition, GAS6 decreased the mRNA content of Mafa. CONCLUSIONS/INTERPRETATION We propose a role for GAS6 in the regulation of pancreatic beta cells in the critical period around the time of birth. Our results support the hypothesis that the reduced beta cell mass seen in LP offspring is caused by a change in the intra-uterine environment that favours premature maturation of the beta cells.
Collapse
Affiliation(s)
- T N Haase
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Panum Institute, Blegdamsvej 3b, Building 6.5, 2200 Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
50
|
Kim Y, Fiel MI, Albanis E, Chou HI, Zhang W, Khitrov G, Friedman SL. Anti-fibrotic activity and enhanced interleukin-6 production by hepatic stellate cells in response to imatinib mesylate. Liver Int 2012; 32:1008-17. [PMID: 22507133 PMCID: PMC3370152 DOI: 10.1111/j.1478-3231.2012.02806.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 03/14/2012] [Indexed: 02/13/2023]
Abstract
OBJECTIVE To examine imatinib mesylate's effects on stellate cell responses in vivo and in vitro. The hepatic stellate cell (HSC) is a key target of anti-fibrotic therapies. Imatinib mesylate is a small molecule receptor tyrosine kinase inhibitor indicated for treatment of chronic myelogenous leukaemia and GI stromal tumours. DESIGN Because imatinib inhibits β-PDGFR signalling, which stimulates HSC proliferation, we assessed its activity in culture and in vivo, and examined downstream targets in a human stellate cell line (LX-2) using cDNA microarray. METHODS AND RESULTS Imatinib inhibited proliferation of LX-2 cells (0.5-10 mM) but not primary human stellate cells, with no effect on viability, associated with attenuated β-PDGFR phosphorylation. Mitochondrial activity and superoxide anion production were decreased in response to imatinib. cDNA microarray uncovered up-regulation of 29 genes in response to imatinib, including interleukin-6 (IL-6) mRNA, which was correlated with progressive IL-6 secretion. Imatinib also decreased gene expression of collagen α(1) (I), alpha smooth muscle actin, β-PDGFR, transforming growth factor β receptor type 1, matrix metalloproteinase 2 and tissue inhibitor of metalloproteinase 2. In vivo, imatinib administered to rats beginning 4 weeks after starting thioacetamide (TAA) led to reduced collagen content, with significant reductions in portal pressure and down-regulation of fibrogenic genes in whole liver. Importantly, hepatic IL-6 mRNA levels were significantly increased in TAA-treated animals receiving imatinib. CONCLUSIONS These findings reinforce the anti-fibrotic activity of imatinib and uncover an unexpected link between inhibition of HSC activation by imatinib and enhanced secretion of IL-6, a regenerative cytokine.
Collapse
Affiliation(s)
- Youngchul Kim
- Division of Liver Diseases, Department of Medicine, The Mount Sinai School of Medicine, New York, NY,College of Oriental Medicine, Kyung Hee University, Seoul, Korea
| | - Maria Isabel Fiel
- Department of Pathology, The Mount Sinai School of Medicine, New York, NY
| | - Efsevia Albanis
- Division of Liver Diseases, Department of Medicine, The Mount Sinai School of Medicine, New York, NY
| | - Hsin I Chou
- Division of Liver Diseases, Department of Medicine, The Mount Sinai School of Medicine, New York, NY
| | - Weijia Zhang
- Bioinformatics Laboratory, Department of Medicine, The Mount Sinai School of Medicine, New York, NY
| | - Gregory Khitrov
- Life Sciences Technology Laboratory Department of Medicine, The Mount Sinai School of Medicine, New York, NY
| | - Scott L. Friedman
- Division of Liver Diseases, Department of Medicine, The Mount Sinai School of Medicine, New York, NY
| |
Collapse
|