1
|
EGFR-TNFR1 pathway in endothelial cell facilitates acute lung injury by NF-κB/MAPK-mediated inflammation and RIP3-dependent necroptosis. Int Immunopharmacol 2023; 117:109902. [PMID: 36827922 DOI: 10.1016/j.intimp.2023.109902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/03/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023]
Abstract
Tumor necrosis factor-α (TNFα) has emerged as a pivotal effector critically correlated with disease severity in acute lung injury (ALI). Because both the excessive activation of epidermal growth factor receptor (EGFR) and tumor necrosis factor receptor 1 (TNFR1) in sepsis-induced vasculitis are markedly diminished through EGFR tyrosine kinase inhibitor, a specific mechanism must exist to modulate TNFR1 cellular fates regulated by EGFR. Here, we demonstrated that EGFR, a specific binding partner of TNFR1, exhibited an increased NF-κB/MAPK-mediated inflammation that was governed by enhanced recruitment of TNFR-associated factor 2 (TRAF2) to TNFR1 complex I in endothelial cell (EC). Moreover, EGFR activation triggered a remarkable increase in the phosphorylation of receptor-interacting protein 1 (RIP1) and its binding with receptor-interacting protein 3 (RIP3) which led to enhanced frequency of necroptosis in complex IIb. Inhibiting the kinase of EGFR disrupted the formation of complex I and complex IIb and prevents EC from NF-κB/MAPK-mediated inflammation and RIP3-dependent necroptosis. Consistently, pharmacological inhibition of EGFR can limit the destructive effects of neutrophils activation and the hyperpermeability of lung vascular in hyperinflammation period. Collectively, we have identified EC-EGFR as a modulator of TNFR1-mediated inflammation and RIP3-dependent necroptosis, providing a possible explanation for the immunological basis of anti-EGFR therapy in sepsis-induced ALI.
Collapse
|
2
|
Clemente L, Bird IM. The epidermal growth factor receptor in healthy pregnancy and preeclampsia. J Mol Endocrinol 2023; 70:e220105. [PMID: 36197759 PMCID: PMC9742168 DOI: 10.1530/jme-22-0105] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 10/05/2022] [Indexed: 11/09/2022]
Abstract
The epidermal growth factor receptor (EGFR) is expressed robustly in the placenta, and critical processes of pregnancy such as placental growth and trophoblast fusion are dependent on EGFR function. However, the role that aberrant EGFR signaling might play in the etiology and/or maintenance of preeclampsia (PE) remains largely unexplored. Recently, we have shown that overexpression of EGFR in cultured uterine artery endothelial cells (UAEC), which express little endogenous EGFR, remaps responsiveness away from vascular endothelial growth factor receptor (VEGFR) signaling and toward EGFR, suggesting that endothelial EGFR expression may be kept low to preserve VEGFR control of angiogenesis. Here we will consider the evidence for the possibility that the endothelial dysfunction observed in PE might in some cases result from elevation of endothelial EGFR. During pregnancy, trophoblasts are known to synthesize large amounts of EGFR protein, and the placenta regularly releases syncytiotrophoblast-derived exosomes and microparticles into the maternal circulation. Although there are no reports of elevated EGFR gene expression in preeclamptic endothelial cells, the ongoing shedding of placental vesicles into the vascular system raises the possibility that EGFR-rich vesicles might fuse with endothelium, thereby contributing to the symptoms of PE by interrupting angiogenesis and blocking pregnancy-adapted vasodilatory function.
Collapse
Affiliation(s)
- Luca Clemente
- Perinatal Research Laboratories, Department of Obstetrics and Gynecology, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53715, USA
| | - Ian M. Bird
- Perinatal Research Laboratories, Department of Obstetrics and Gynecology, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53715, USA
- Department of Pediatrics, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53715, USA
| |
Collapse
|
3
|
Li X, Ge J, Li M, Deng S, Li J, Ma Y, Zhang J, Zheng Y, Ma L. Network pharmacology, molecular docking technology integrated with pharmacodynamic study to reveal the potential targets of Schisandrol A in drug-induced liver injury by acetaminophen. Bioorg Chem 2021; 118:105476. [PMID: 34788696 DOI: 10.1016/j.bioorg.2021.105476] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 07/02/2021] [Accepted: 11/04/2021] [Indexed: 12/12/2022]
Abstract
Schisandrae Chinensis Fructus (SCF) was a Traditional Chinese Medicine for protecting liver. However, underlying therapeutic mechanisms of these bioactive lignans from SCF similar hepatoprotective effects against drug-induced liver injury (DILI) by acetaminophen (APAP) are still unclear. This study aims to discover the potential regulation mechanisms of Schisandrol A in the treatment of DILI by APAP. The integrated UPLC-Q-TOF/MS, pharmacodynamic study, histopathological combination with network pharmacology and molecular docking technology were used to explore the potential mechanisms. The results showed that Schisandrol A reduced the level of AST, ALT, MDA, PNP, TNF-α and IL-1β, increased the levels of the GSH against acute liver failure. Additionally, Schisandrol A could improve the morphological characteristics of DILI by APAP in mice with liver tissue. Molecular docking results had showed that Schisandrol A with high scores when docking with COX-2, ALOX5, CYP2E1, CYP2C9, CYP2C19, EGFR SRC, Nrf2, MAPK14 and MAPK8. The study demonstrated that Schisandrol A could play critical roles in DILI by APAP via regulating TNF signaling pathway, inhibiting oxidative stress, inflammation and inhibiting the activities of cytochrome P450 enzymes, which contributed to searching for leading compounds and the development of new drugs for DILI by APAP.
Collapse
Affiliation(s)
- Xiankuan Li
- Tianjin University of Traditional Chinese Medicine, 301617, PR China
| | - Jiaming Ge
- Tianjin University of Traditional Chinese Medicine, 301617, PR China
| | - Mengyuan Li
- Tianjin University of Traditional Chinese Medicine, 301617, PR China
| | - Sha Deng
- Tianjin University of Traditional Chinese Medicine, 301617, PR China
| | - Jiarong Li
- Tianjin University of Traditional Chinese Medicine, 301617, PR China
| | - Yucong Ma
- Changchun University of Chinese Medicine, Changchun, 130117, PR China
| | - Jian Zhang
- Tianjin University of Traditional Chinese Medicine, 301617, PR China
| | - Yanchao Zheng
- Tianjin University of Traditional Chinese Medicine, 301617, PR China.
| | - Lin Ma
- Tianjin University of Traditional Chinese Medicine, 301617, PR China.
| |
Collapse
|
4
|
Kemeny NE, Chou JF, Capanu M, Chatila WK, Shi H, Sanchez-Vega F, Kingham TP, Connell LC, Jarnagin WR, D'Angelica MI. A Randomized Phase II Trial of Adjuvant Hepatic Arterial Infusion and Systemic Therapy With or Without Panitumumab After Hepatic Resection of KRAS Wild-type Colorectal Cancer. Ann Surg 2021; 274:248-254. [PMID: 33938493 PMCID: PMC9351589 DOI: 10.1097/sla.0000000000004923] [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] [Indexed: 11/26/2022]
Abstract
OBJECTIVE/BACKGROUND The purpose was to determine whether adding Pmab versus no Pmab to an adjuvant regimen of hepatic arterial infusion (HAI) of floxuridine (FUDR) plus systemic (SYS) leucovorin, fluorouracil, and irinotecan (FOLFIRI) improves 15-month recurrence-free survival for patients with RAS wild-type colorectal cancer. Secondary endpoints included overall survival, toxicity, and influence of predictive biomarkers. METHODS This phase II trial randomized patients with KRAS wild-type resected colorectal liver metastases to adjuvant HAI FUDR + SYS FOLFIRI +/- Pmab (NCT01312857). Patients were stratified by clinical risk score and previous chemotherapy. Based on an exact binomial design, if one arm had ≥24 patients alive and disease-free at 15 months that regimen was considered promising for further investigation. RESULTS Seventy-five patients were randomized. Patient characteristics and toxicity were not different in the 2 arms, except for rash in +Pmab arm. Grade 3/4 elevation in bilirubin or alkaline phosphatase did not differ in the 2 arms. Twenty-five (69%; 95% CI, 53-82) patients in the Pmab arm versus 18 (47%; 95% CI, 32-63) patients in the arm without Pmab were alive and recurrence-free at 15 months. Only the Pmab arm met the decision rule, while the other arm did not. After median follow-up of 56.6 months, 3-year recurrence-free survival was 57% (95% CI, 43-76) and 42% (95% CI, 29-61), and 3-year overall survival was 97% (95% CI, 90-99) and 91% (95% CI, 83-99), +/- Pmab, respectively. CONCLUSIONS The addition of Pmab to HAI FUDR + SYS FOLFIRI showed promising activity without increased biliary toxicity and should be further investigated in a larger trial.
Collapse
Affiliation(s)
- Nancy E Kemeny
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Joanne F Chou
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marinela Capanu
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Walid K Chatila
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Hongyu Shi
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Francisco Sanchez-Vega
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Thomas Peter Kingham
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Louise Catherine Connell
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - William R Jarnagin
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael I D'Angelica
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
5
|
Théret N, Bouezzeddine F, Azar F, Diab-Assaf M, Legagneux V. ADAM and ADAMTS Proteins, New Players in the Regulation of Hepatocellular Carcinoma Microenvironment. Cancers (Basel) 2021; 13:cancers13071563. [PMID: 33805340 PMCID: PMC8037375 DOI: 10.3390/cancers13071563] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Members of the adamalysin family are multi-domain proteins involved in many cancer-related functions. In this review, we will examine the literature on the involvement of adamalysins in hepatocellular carcinoma progression and their importance in the tumor microenvironment where they regulate the inflammatory response and the epithelial–mesenchymal transition. We complete this review with an analysis of adamalysin expression in a large cohort of patients with hepatocellular carcinoma from The Cancer Genome Atlas (TCGA) database. These original results give a new insight into the involvement of all adamalysins in the primary liver cancer. Abstract The tumor microenvironment plays a major role in tumor growth, invasion and resistance to chemotherapy, however understanding how all actors from microenvironment interact together remains a complex issue. The tumor microenvironment is classically represented as three closely connected components including the stromal cells such as immune cells, fibroblasts, adipocytes and endothelial cells, the extracellular matrix (ECM) and the cytokine/growth factors. Within this space, proteins of the adamalysin family (ADAM for a disintegrin and metalloproteinase; ADAMTS for ADAM with thrombospondin motifs; ADAMTSL for ADAMTS-like) play critical roles by modulating cell–cell and cell–ECM communication. During last decade, the implication of adamalysins in the development of hepatocellular carcinoma (HCC) has been supported by numerous studies however the functional characterization of most of them remain unsettled. In the present review we propose both an overview of the literature and a meta-analysis of adamalysins expression in HCC using data generated by The Cancer Genome Atlas (TCGA) Research Network.
Collapse
Affiliation(s)
- Nathalie Théret
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en santé, Environnement et Travail)-UMR_S1085, University of Rennes 1, 35000 Rennes, France; (F.A.); (V.L.)
- Correspondence:
| | - Fidaa Bouezzeddine
- Molecular Cancer and Pharmaceutical Biology Laboratory, Faculty of Sciences II, Lebanese University Fanar, 1500 Beirut, Lebanon; (F.B.); (M.D.-A.)
| | - Fida Azar
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en santé, Environnement et Travail)-UMR_S1085, University of Rennes 1, 35000 Rennes, France; (F.A.); (V.L.)
| | - Mona Diab-Assaf
- Molecular Cancer and Pharmaceutical Biology Laboratory, Faculty of Sciences II, Lebanese University Fanar, 1500 Beirut, Lebanon; (F.B.); (M.D.-A.)
| | - Vincent Legagneux
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en santé, Environnement et Travail)-UMR_S1085, University of Rennes 1, 35000 Rennes, France; (F.A.); (V.L.)
| |
Collapse
|
6
|
Abolfathi H, Sheikhpour M, Shahraeini SS, Khatami S, Nojoumi SA. Studies in lung cancer cytokine proteomics: a review. Expert Rev Proteomics 2021; 18:49-64. [PMID: 33612047 DOI: 10.1080/14789450.2021.1892491] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Proteins are molecules that have role in the progression of the diseases. Proteomics is a tool that can play an effective role in identifying diagnostic and therapeutic biomarkers for lung cancer. Cytokines are proteins that play a decisive role in activating body's immune system in lung cancer. They can increase the growth of the tumor (oncogenic cytokines) or limit tumor growth (anti-tumor cytokines) by regulating related signaling pathways such as proliferation, growth, metastasis, and apoptosis. AREAS COVERED In the present study, a total of 223 papers including 196 research papers and 27 review papers, extracted from PubMed and Scopus and published from 1997 to present, are reviewed. The most important involved-cytokines in lung cancer including TNF-α, IFN- γ, TGF-β, VEGF and interleukins such as IL-6, IL-17, IL-8, IL-10, IL-22, IL-1β and IL-18 are introduced. Also, the pathological and biological role of such cytokines in cancer signaling pathways is explained. EXPERT OPINION In lung cancer, the cytokine expression changes under the physiological conditions of the immune system, and inflammatory cytokines are associated with the progression of lung cancer. Therefore, the cytokine expression profile can be used in the diagnosis, prognosis, prediction of therapeutic responses, and survival of patients with lung cancer.
Collapse
Affiliation(s)
- Hanie Abolfathi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Mojgan Sheikhpour
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Sadegh Shahraeini
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Shohreh Khatami
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Ali Nojoumi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
7
|
Pathophysiology of Lung Disease and Wound Repair in Cystic Fibrosis. PATHOPHYSIOLOGY 2021; 28:155-188. [PMID: 35366275 PMCID: PMC8830450 DOI: 10.3390/pathophysiology28010011] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 12/11/2022] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive, life-threatening condition affecting many organs and tissues, the lung disease being the chief cause of morbidity and mortality. Mutations affecting the CF Transmembrane Conductance Regulator (CFTR) gene determine the expression of a dysfunctional protein that, in turn, triggers a pathophysiological cascade, leading to airway epithelium injury and remodeling. In vitro and in vivo studies point to a dysregulated regeneration and wound repair in CF airways, to be traced back to epithelial CFTR lack/dysfunction. Subsequent altered ion/fluid fluxes and/or signaling result in reduced cell migration and proliferation. Furthermore, the epithelial-mesenchymal transition appears to be partially triggered in CF, contributing to wound closure alteration. Finally, we pose our attention to diverse approaches to tackle this defect, discussing the therapeutic role of protease inhibitors, CFTR modulators and mesenchymal stem cells. Although the pathophysiology of wound repair in CF has been disclosed in some mechanisms, further studies are warranted to understand the cellular and molecular events in more details and to better address therapeutic interventions.
Collapse
|
8
|
Nakayama I, Higa-Nakamine S, Uehara A, Sugahara K, Kakinohana M, Yamamoto H. Regulation of epidermal growth factor receptor expression and morphology of lung epithelial cells by interleukin-1β. J Biochem 2021; 168:113-123. [PMID: 32016419 DOI: 10.1093/jb/mvaa015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 01/16/2020] [Indexed: 12/31/2022] Open
Abstract
Accumulating evidences suggested that the overactivation of epidermal growth factor receptor (EGFR) was involved in the development of adult respiratory distress syndrome and pulmonary fibrosis. Elucidation of the mechanisms that regulate EGFR residence on the plasma membrane during inflammatory lung conditions is important for identifying potential therapies. We have demonstrated that flagellin phosphorylated EGFR at Ser1047 and induced transient EGFR internalization. In this study, we examined the molecular pathway and effect of interleukin 1 beta (IL-1β) on EGFR in alveolar epithelial cells. Treatment of A549 cells with IL-1β induced the activation of p38 mitogen-activated protein kinase (MAP kinase) and MAP kinase-activated protein kinase-2 (MAPKAPK-2), as well as EGFR phosphorylation at serine 1047. Both MAPKAPK-2 activation and EGFR phosphorylation were inhibited by SB203580, a p38 MAP kinase inhibitor. In addition, MK2a inhibitor (a MAPKAPK-2 inhibitor) suppressed EGFR phosphorylation. Assessment of the biotinylation of cell surface proteins indicated that IL-1β induced EGFR internalization. Furthermore, long-term treatment of A549 cells with IL-1β caused morphological changes and loss of cell-cell contact. Moreover, IL-1β augmented the effect of transforming growth factor beta 1 on the epithelial-mesenchymal transition. These results suggested that IL-1β regulates EGFR functions and induces morphological changes of alveolar epithelial cells.
Collapse
Affiliation(s)
- Izumi Nakayama
- Department of Biochemistry, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.,Intensive Care Unit, Department of Internal Medicine, Okinawa Chubu Hospital, Okinawa 904-2293, Japan
| | - Sayomi Higa-Nakamine
- Department of Biochemistry, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Ayako Uehara
- Department of Anesthesiology, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Kazuhiro Sugahara
- Department of Anesthesiology, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Manabu Kakinohana
- Department of Anesthesiology, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Hideyuki Yamamoto
- Department of Biochemistry, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| |
Collapse
|
9
|
Mercogliano MF, Bruni S, Mauro F, Elizalde PV, Schillaci R. Harnessing Tumor Necrosis Factor Alpha to Achieve Effective Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13030564. [PMID: 33540543 PMCID: PMC7985780 DOI: 10.3390/cancers13030564] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/17/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor alpha (TNFα) is a pleiotropic cytokine known to have contradictory roles in oncoimmunology. Indeed, TNFα has a central role in the onset of the immune response, inducing both activation and the effector function of macrophages, dendritic cells, natural killer (NK) cells, and B and T lymphocytes. Within the tumor microenvironment, however, TNFα is one of the main mediators of cancer-related inflammation. It is involved in the recruitment and differentiation of immune suppressor cells, leading to evasion of tumor immune surveillance. These characteristics turn TNFα into an attractive target to overcome therapy resistance and tackle cancer. This review focuses on the diverse molecular mechanisms that place TNFα as a source of resistance to immunotherapy such as monoclonal antibodies against cancer cells or immune checkpoints and adoptive cell therapy. We also expose the benefits of TNFα blocking strategies in combination with immunotherapy to improve the antitumor effect and prevent or treat adverse immune-related effects.
Collapse
Affiliation(s)
- María Florencia Mercogliano
- Laboratorio de Biofisicoquímica de Proteínas, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales-Consejo Nacional de Investigaciones Científicas y Técnicas (IQUIBICEN-CONICET), Buenos Aires 1428, Argentina;
| | - Sofía Bruni
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
| | - Florencia Mauro
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
| | - Patricia Virginia Elizalde
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
| | - Roxana Schillaci
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
- Correspondence: ; Tel.: +54-11-4783-2869; Fax: +54-11-4786-2564
| |
Collapse
|
10
|
Resina Draconis Reduces Acute Liver Injury and Promotes Liver Regeneration after 2/3 Partial Hepatectomy in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:2305784. [PMID: 33082819 PMCID: PMC7563078 DOI: 10.1155/2020/2305784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/17/2020] [Accepted: 09/07/2020] [Indexed: 12/29/2022]
Abstract
Aim To investigate the protective effects and possible mechanisms of action of resina draconis (RD) on acute liver injury and liver regeneration after 2/3 partial hepatectomy (PH) in mice. Methods 2/3 PH was used to induce acute liver injury. Mice were divided into three groups: sham, vehicle + 2/3 PH, and RD + 2/3 PH. Resina draconis was administered intragastrically after 2/3 PH into the RD + 2/3 PH group, and the same volume of vehicle (1% sodium carboxymethyl cellulose) was injected into the vehicle + 2/3 PH group and sham group mice. The index of liver to body weight (ILBW) and proliferating cell nuclear antigen (PCNA) were assayed to evaluate liver regeneration. Blood and liver tissues were collected for serological and western blotting analysis. Results Resina draconis protected against 2/3 PH-induced acute severe liver injury and promoted liver regeneration as shown by significantly increased ILBW compared with that of controls. 2/3 PH increased serum AST and ALT levels, which were significantly decreased by RD treatment, while 2/3 PH decreased serum TP and ALB, which were increased by RD treatment. In the RD + 2/3 PH group, PCNA expression was significantly increased compared with the 2/3 PH group. Further, hepatocyte growth factor (HGF), TNFα, and EGFR levels were increased in the RD group at postoperative days 2 and 4 compared with the those in the 2/3 PH group. Conclusion Our results suggest that RD ameliorates acute hepatic injury and promotes liver cell proliferation, liver weight restoration, and liver function after 2/3 PH, probably via HGF, TNFα, and EGFR signaling.
Collapse
|
11
|
Della-Fazia MA, Castelli M, Piobbico D, Pieroni S, Servillo G. The Ins and Outs of HOPS/TMUB1 in biology and pathology. FEBS J 2020; 288:2773-2783. [PMID: 32860479 DOI: 10.1111/febs.15539] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/28/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022]
Abstract
Liver regeneration represents an outstanding tool to study not only proliferation, but also other important processes such as inflammation, regenerative response or stem cell biology. Several novel genes have been identified as being involved in the proliferation of residual hepatocytes. One of them, HOPS/TMUB1, is proving to be a significant player in the control of proliferation, both contributing to genomic stability and as a partner of essential molecules. HOPS is an ubiquitin-like protein, shuttling from nucleus to cytoplasm, and it is engaged in a number of biological and physiopathological functions. HOPS overexpression in tumour cell lines strongly reduces proliferation, arresting cell cycle in G0 /G1 . HOPS is involved in centrosome assembly and maintenance, and its knockdown causes genomic instability. Moreover, a direct interaction of HOPS with nucleophosmin (NPM) and p19Arf has been established, resulting in proper control of p19Arf stability and localization. These data indicate that HOPS acts as a functional bridge in the interaction between NPM and p19Arf , providing new mechanistic insight into how NPM and p19Arf will oppose cell proliferation. HOPS exerts a control in p53 stability, directing p53 mitochondrial apoptosis and cytoplasmic localization. HOPS plays a direct role as novel post-translational modifier of p53, much like SUMO or NEDD. HOPS is overexpressed in a high number of human tumours in patients affected by large intestinal, CNS, liver and oesophageal tumours. This review highlights HOPS involvement in distinct cellular functions, establishing its role as a key player in cell biology and pathology in a broader context.
Collapse
Affiliation(s)
| | | | - Danilo Piobbico
- Department of Experimental Medicine, University of Perugia, Italy
| | - Stefania Pieroni
- Department of Experimental Medicine, University of Perugia, Italy
| | | |
Collapse
|
12
|
Clemente L, Boeldt DS, Grummer MA, Morita M, Morgan TK, Wiepz GJ, Bertics PJ, Bird IM. Adenoviral transduction of EGFR into pregnancy-adapted uterine artery endothelial cells remaps growth factor induction of endothelial dysfunction. Mol Cell Endocrinol 2020; 499:110590. [PMID: 31550517 PMCID: PMC6886699 DOI: 10.1016/j.mce.2019.110590] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 09/19/2019] [Accepted: 09/19/2019] [Indexed: 12/13/2022]
Abstract
During pregnancy, uterine vascular vasodilation is enhanced through adapted Ca2+ signaling, facilitated through increased endothelial connexin 43 (Cx43) gap junctional communication (GJC). In preeclampsia (PE), this adaptive response is missing. Of note, the angiogenic factor VEGF can also act via Src and ERK to close Cx43 gap junctions. While VEGFR2 is necessary for such closure, a role VEGFR1 is less clear. We reasoned if VEGFR2 is acting alone, then substituting another growth factor receptor with VEGFR2-like signaling should have the same effect. In uterine artery endothelial cells derived from pregnant sheep (P-UAEC), endogenous EGFR expression is very low. When we used adenovirus to raise EGFR, we also dose-dependently induced EGF-sensitive Cx43 phosphorylation mainly via ERK, and corresponding loss of Ca2+ bursts, but eliminated VEGF effects on phosphorylation of Cx43 or loss of Ca2+ bursting. This surprising observation suggests that while activated EGFR may indeed substitute for VEGFR2, it also sequesters a limited pool of effector molecules needed for VEGFR2 to phosphorylate Cx43. Thus, low endogenous EGFR expression in P-UAEC may be a necessary strategy to allow VEGFR-2 control of GJC, a first step in initiating angiogenesis in healthy pregnancy. Of further note, trophoblasts are rich in EGFR, and we have demonstrated shed PLAP+/EGFR + extracellular vesicles in maternal circulation in first trimester plasma samples using nanoscale high resolution flow cytometry. Collectively our data suggest that placenta derived exosomes positive for EGFR should be further considered as a possible cause of endothelial dysfunction in women with PE.
Collapse
Affiliation(s)
- Luca Clemente
- Perinatal Research Laboratories, Department of Obstetrics and Gynecology, University of Wisconsin - Madison, School of Medicine and Public Health, Madison, WI, 53715, USA
| | - Derek S Boeldt
- Perinatal Research Laboratories, Department of Obstetrics and Gynecology, University of Wisconsin - Madison, School of Medicine and Public Health, Madison, WI, 53715, USA
| | - Mary A Grummer
- Perinatal Research Laboratories, Department of Obstetrics and Gynecology, University of Wisconsin - Madison, School of Medicine and Public Health, Madison, WI, 53715, USA
| | - Mayu Morita
- Departments of Pathology, Obstetrics & Gynecology, and Biomedical Engineering, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Terry K Morgan
- Departments of Pathology, Obstetrics & Gynecology, and Biomedical Engineering, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Greg J Wiepz
- Department of Comparative Biosciences, University of Wisconsin - Madison, School of Veterinary Medicine, Madison, WI, 53715, USA
| | - Paul J Bertics
- Department of Biomolecular Chemistry, University of Wisconsin - Madison, School of Medicine and Public Health, Madison, WI, 53715, USA
| | - Ian M Bird
- Perinatal Research Laboratories, Department of Obstetrics and Gynecology, University of Wisconsin - Madison, School of Medicine and Public Health, Madison, WI, 53715, USA; Department of Pediatrics, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA.
| |
Collapse
|
13
|
Tanner MA, Thomas TP, Grisanti LA. Death receptor 5 contributes to cardiomyocyte hypertrophy through epidermal growth factor receptor transactivation. J Mol Cell Cardiol 2019; 136:1-14. [PMID: 31473246 DOI: 10.1016/j.yjmcc.2019.08.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 12/30/2022]
Abstract
Cardiomyocyte survival and death contributes to many cardiac diseases. A common mechanism of cardiomyocyte death is through apoptosis however, numerous death receptors (DR) have been virtually unstudied in the context of cardiovascular disease. Previous studies have identified TNF-related apoptosis inducing ligand (TRAIL) and its receptor, DR5, as being altered in a chronic catecholamine administration model of heart failure, and suggest a role of non-canonical signaling in cardiomyocytes. Furthermore, multiple clinical studies have identified TRAIL or DR5 as biomarkers in the prediction of severity and mortality following myocardial infarction and in heart failure development risk suggesting a role of DR5 signaling in the heart. While TRAIL/DR5 have been extensively studied as a potential cancer therapeutic due to their ability to selectively activate apoptosis in cancer cells, TRAIL and DR5 are highly expressed in the heart where their function is uncharacterized. However, many non-transformed cell types are resistant to TRAIL-induced apoptosis suggesting non-canonical functions in non-cancerous cell types. Our goal was to determine the role of DR5 in the heart with the hypothesis that DR5 does not induce cardiomyocyte apoptosis but initiates non-canonical signaling to promote cardiomyocyte growth and survival. Histological analysis of hearts from mice treated with a DR5 agonists showed increased hypertrophy with no differences in cardiomyocyte death, fibrosis or function. Mechanistic studies in the heart and isolated cardiomyocytes identified ERK1/2 activation with DR5 agonist treatment which contributed to hypertrophy. Furthermore, epidermal growth factor receptor (EGFR) was activated following DR5 agonist treatment through activation of MMP and HB-EGFR cleavage and specific inhibitors of MMP and EGFR prevented DR5-mediated ERK1/2 signaling and hypertrophy. Taken together, these studies identify a previously unidentified role for DR5 in the heart, which does not promote apoptosis but acts through non-canonical MMP-EGFR-ERK1/2 signaling mechanisms to contribute to cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Miles A Tanner
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Toby P Thomas
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Laurel A Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
14
|
Riddiough GE, Fifis T, Muralidharan V, Perini MV, Christophi C. Searching for the link; mechanisms underlying liver regeneration and recurrence of colorectal liver metastasis post partial hepatectomy. J Gastroenterol Hepatol 2019; 34:1276-1286. [PMID: 30828863 DOI: 10.1111/jgh.14644] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/23/2019] [Accepted: 02/28/2019] [Indexed: 12/13/2022]
Abstract
Despite excellent treatment of primary colorectal cancer, the majority of deaths occur as a result of metastasis to the liver. Recent population studies have estimated that one quarter of patients with colorectal cancer will incur synchronous or metachronous colorectal liver metastasis. However, only one quarter of these patients will be eligible for potentially curative resection. Tumor recurrence occurs in reportedly 60% of patients undergoing hepatic resection, and the majority of intrahepatic recurrence occurs within the first 6 months of surgery. The livers innate ability to restore its homeostatic size, and volume facilitates major hepatic resection that currently offers the only chance of cure to patients with extensive hepatic metastases. Experimental and clinical evidence supports the notion that following partial hepatectomy, liver regeneration (LR) paradoxically drives tumor progression and increases the risk of recurrence. It is becoming increasingly clear that the processes that drive liver organogenesis, regeneration, and tumor progression are inextricably linked. This presents a major hurdle in the management of colorectal liver metastasis and other hepatic malignancies because therapies that reduce the risk of recurrence without hampering LR are sought. The processes and pathways underlying these phenomena are multiple, complex, and cross-communicate. In this review, we will summarize the common mechanisms contributing to both LR and tumor recurrence.
Collapse
Affiliation(s)
- Georgina E Riddiough
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Theodora Fifis
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | | | - Marcos V Perini
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Christopher Christophi
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| |
Collapse
|
15
|
Abstract
The liver has a unique ability of regenerating after injuries or partial loss of its mass. The mechanisms responsible for liver regeneration - mostly occurring when the hepatic tissue is damaged or functionally compromised by metabolic stress - have been studied in considerable detail over the last few decades, because this phenomenon has both basic-biology and clinical relevance. More specifically, recent interest has been focusing on the widespread occurrence of abnormal nutritional habits in the Western world that result in an increased prevalence of non-alcoholic fatty liver disease (NAFLD). NAFLD is closely associated with insulin resistance and dyslipidemia, and it represents a major clinical challenge. The disease may progress to steatohepatitis with persistent inflammation and progressive liver damage, both of which will compromise regeneration under conditions of partial hepatectomy in surgical oncology or in liver transplantation procedures. Here, we analyze the impact of ER stress and SIRT1 in lipid metabolism and in fatty liver pathology, and their consequences on liver regeneration. Moreover, we discuss the fine interplay between ER stress and SIRT1 functioning when contextualized to liver regeneration. An improved understanding of the cellular and molecular intricacies contributing to liver regeneration could be of great clinical relevance in areas as diverse as obesity, metabolic syndrome and type 2 diabetes, as well as oncology and transplantation.
Collapse
Affiliation(s)
| | - Giuseppe Servillo
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
16
|
Ulmer TF, Fragoulis A, Dohmeier H, Kroh A, Andert A, Stoppe C, Alizai H, Klink C, Coburn M, Neumann UP. Argon Delays Initiation of Liver Regeneration after Partial Hepatectomy in Rats. Eur Surg Res 2017; 58:204-215. [PMID: 28433997 DOI: 10.1159/000466690] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 02/28/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND The liver can heal up to restitutio ad integrum following damage resulting from various causes. Different studies have demonstrated the protective effect of argon on various cells and organs. To the best of our knowledge, the organ-protective effects of the noble gas argon on the liver have not yet been investigated, although argon appears to influence signal paths that are well-known mediators of liver regeneration. We hypothesized that argon inhalation prior to partial hepatectomy (70%) has a positive effect on the initiation of liver regeneration in rats. METHODS Partial hepatectomy (70%) with or without inhaled argon (50 vol%) was performed for 1 h. Liver tissue was harvested after 3, 36, and 96 h to analyze the mRNA and protein expression of hepatocyte growth factor (HGF), interleukin-6 (IL-6), tumor necrosis factor-α, and extracellular signal-regulated kinase 1/2. Histological tissue samples were prepared for immunohistochemistry (bromodeoxyuridine [BrdU], Ki-67, and TUNEL) and blood was analyzed regarding the effects of argon on liver function. Statistical analyses were performed using 1-way ANOVA followed by the post hoc Tukey-Kramer test. RESULTS After 3 h, the primary outcome parameter of hepatocyte proliferation was significantly reduced with argon 50 vol% inhalation in comparison to nitrogen inhalation (BrdU: 15.7 ± 9.7 vs. 7.7 ± 3.1 positive cells/1,000 hepatocytes, p = 0.013; Ki-67: 17.6 ± 13.3 vs. 4.7 ± 5.4 positive cells/1,000 hepatocytes, p = 0.006). This was most likely mediated by significant downregulation of HGF (after 3 h: 5.2 ± 3.2 vs. 2.3 ± 1.0 fold, p = 0.032; after 96 h: 2.1 ± 0.5 vs. 1.3 ± 0.3 fold, p = 0.029) and IL-6 (after 3 h: 43.7 ± 39.6 vs. 8.5 ± 9.2 fold, p = 0.032). Nevertheless, we could detect no significant effect on the weight of the residual liver, liver-body weight ratio, or liver blood test results after argon inhalation. CONCLUSION Impairment of liver regeneration was apparent after argon 50 vol% inhalation that was most probably mediated by downregulation of HGF and IL-6 in the initial phase. However, the present study was not adequately powered to prove that argon has detrimental effects on the liver. Further studies are needed to evaluate the effects of argon on livers with preexisting conditions as well as on ischemia-reperfusion models.
Collapse
Affiliation(s)
- Tom Florian Ulmer
- Department of General, Visceral, and Transplantation Surgery, University Hospital of RWTH Aachen, Aachen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Wang M, Xie R, Si H, Shen B. Integrated bioinformatics analysis of miRNA expression in osteosarcoma. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:936-943. [PMID: 27315542 DOI: 10.1080/21691401.2016.1196456] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The expression of miRNA influencing the pathogenesis of OS have been reported previously, however, different samples selection and sequencing platforms made obvious differences in miRNA expression analysis. We aim to identify reliable prognostic and treatment biomarkers for OS by systematic analysis of miRNAs expression data sets from biased data set. Seven miRNA data sets were selected from corresponding articles. Collectively, two miRNAs, hsa-miR-19-3p and hsa-miR106b-3p, and transcription factor SIX3 were identified and may be reliable markers for prognostic and treatment of osteosarcoma.
Collapse
Affiliation(s)
- Maoyuan Wang
- a West China School of Medicine, Sichuan University , Chengdu , China
| | - Ruilian Xie
- b Department of Medical Oncology , The First Affiliated Hospital of Gannan Medical College , Ganzhou , China
| | - Haibo Si
- c Department of Orthopaedics , West China Hospital, Sichuan University , Chengdu , China
| | - Bin Shen
- c Department of Orthopaedics , West China Hospital, Sichuan University , Chengdu , China
| |
Collapse
|
18
|
Lee JI, Wright JH, Johnson MM, Bauer RL, Sorg K, Yuen S, Hayes BJ, Nguyen L, Riehle KJ, Campbell JS. Role of Smad3 in platelet-derived growth factor-C-induced liver fibrosis. Am J Physiol Cell Physiol 2015; 310:C436-45. [PMID: 26632601 DOI: 10.1152/ajpcell.00423.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 12/01/2015] [Indexed: 12/31/2022]
Abstract
Chronic liver injury leads to fibrosis and cirrhosis. Cirrhosis, the end stage of chronic liver disease, is a leading cause of death worldwide and increases the risk of developing hepatocellular carcinoma. Currently, there is a lack of effective antifibrotic therapies to treat fibrosis and cirrhosis. Development of antifibrotic therapies requires an in-depth understanding of the cellular and molecular mechanisms involved in inflammation and fibrosis after hepatic injury. Two growth factor signaling pathways that regulate liver fibrosis are transforming growth factor-β (TGFβ) and platelet-derived growth factor (PDGF). However, their specific contributions to fibrogenesis are not well understood. Using a genetic model of liver fibrosis, we investigated whether the canonical TGFβ signaling pathway was necessary for fibrogenesis. PDGF-C transgenic (PDGF-C Tg) mice were intercrossed with mice that lack Smad3, and molecular and histological fibrosis was analyzed. PDGF-C Tg mice that also lacked Smad3 had less fibrosis and improved liver lobule architecture. Loss of Smad3 also reduced expression of collagen genes, which were induced by PDGF-C, but not the expression of genes frequently associated with hepatic stellate cell (HSC) activation. In vitro HSCs isolated from Smad3-null mice proliferated more slowly than cells from wild-type mice. Taken together, these findings indicate that PDGF-C activates TGFβ/Smad3 signaling pathways to regulate HSC proliferation, collagen production and ultimately fibrosis. In summary, these results suggest that inhibition of both PDGF and TGFβ signaling pathways may be required to effectively attenuate fibrogenesis in patients with chronic liver disease.
Collapse
Affiliation(s)
- Jung Il Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Jocelyn H Wright
- Department of Pathology, University of Washington, Seattle, Washington; and
| | - Melissa M Johnson
- Department of Pathology, University of Washington, Seattle, Washington; and
| | - Renay L Bauer
- Department of Pathology, University of Washington, Seattle, Washington; and
| | - Kristina Sorg
- Department of Pathology, University of Washington, Seattle, Washington; and
| | - Sebastian Yuen
- Department of Pathology, University of Washington, Seattle, Washington; and
| | - Brian J Hayes
- Department of Pathology, University of Washington, Seattle, Washington; and
| | - Lananh Nguyen
- Department of Pathology, University of Washington, Seattle, Washington; and
| | - Kimberly J Riehle
- Department of Surgery, University of Washington, Seattle, Washington
| | - Jean S Campbell
- Department of Pathology, University of Washington, Seattle, Washington; and
| |
Collapse
|
19
|
Paricalcitol Inhibits Aldosterone-Induced Proinflammatory Factors by Modulating Epidermal Growth Factor Receptor Pathway in Cultured Tubular Epithelial Cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:783538. [PMID: 26064952 PMCID: PMC4438184 DOI: 10.1155/2015/783538] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/11/2015] [Indexed: 11/17/2022]
Abstract
Chronic kidney disease is characterized by Vitamin D deficiency and activation of the renin-angiotensin-aldosterone system. Increasing data show that vitamin D receptor agonists (VDRAs) exert beneficial effects in renal disease and possess anti-inflammatory properties, but the underlying mechanism remains unknown. Emerging evidence suggests that "a disintegrin and metalloproteinase" (ADAM)/epidermal growth factor receptor (EGFR) signalling axis contributes to renal damage. Aldosterone induces EGFR transactivation regulating several processes including cell proliferation and fibrosis. However, data on tubular epithelial cells is scarce. We have found that, in cultured tubular epithelial cells, aldosterone induced EGFR transactivation via TGF-α/ADAM17. Blockade of the TGF-α/ADAM17/EGFR pathway inhibited aldosterone-induced proinflammatory gene upregulation. Moreover, among the potential downstream mechanisms, we found that TGF-α/ADAM17/EGFR inhibition blocked ERK and STAT-1 activation in response to aldosterone. Next, we investigated the involvement of TGF-α/ADAM17/EGFR axis in VDRA anti-inflammatory effects. Preincubation with the VDRA paricalcitol inhibited aldosterone-induced EGFR transactivation, TGF-α/ADAM-17 gene upregulation, and downstream mechanisms, including proinflammatory factors overexpression. In conclusion, our data suggest that the anti-inflammatory actions of paricalcitol in tubular cells could depend on the inhibition of TGF-α/ADAM17/EGFR pathway in response to aldosterone, showing an important mechanism of VDRAs action.
Collapse
|
20
|
Expression of tumor necrosis factor-alpha-mediated genes predicts recurrence-free survival in lung cancer. PLoS One 2014; 9:e115945. [PMID: 25548907 PMCID: PMC4280165 DOI: 10.1371/journal.pone.0115945] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 11/26/2014] [Indexed: 12/25/2022] Open
Abstract
In this study, we conducted a meta-analysis on high-throughput gene expression data to identify TNF-α-mediated genes implicated in lung cancer. We first investigated the gene expression profiles of two independent TNF-α/TNFR KO murine models. The EGF receptor signaling pathway was the top pathway associated with genes mediated by TNF-α. After matching the TNF-α-mediated mouse genes to their human orthologs, we compared the expression patterns of the TNF-α-mediated genes in normal and tumor lung tissues obtained from humans. Based on the TNF-α-mediated genes that were dysregulated in lung tumors, we developed a prognostic gene signature that effectively predicted recurrence-free survival in lung cancer in two validation cohorts. Resampling tests suggested that the prognostic power of the gene signature was not by chance, and multivariate analysis suggested that this gene signature was independent of the traditional clinical factors and enhanced the identification of lung cancer patients at greater risk for recurrence.
Collapse
|
21
|
Karakawa M, Komine M, Hanakawa Y, Tsuda H, Sayama K, Tamaki K, Ohtsuki M. CCL27 Is Downregulated by Interferon Gamma via Epidermal Growth Factor Receptor in Normal Human Epidermal Keratinocytes. J Cell Physiol 2014; 229:1935-45. [DOI: 10.1002/jcp.24643] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 04/02/2014] [Indexed: 01/09/2023]
Affiliation(s)
- Masaru Karakawa
- Department of Dermatology; University of Tokyo; Bunkyo-ku Tokyo Japan
- Department of Dermatology; Jichi Medical University; Shimotsuke Tochigi Japan
| | - Mayumi Komine
- Department of Dermatology; University of Tokyo; Bunkyo-ku Tokyo Japan
- Department of Dermatology; Jichi Medical University; Shimotsuke Tochigi Japan
| | - Yasushi Hanakawa
- Department of Dermatology; University of Ehime; Tou-on-shi Ehime Japan
| | - Hidetoshi Tsuda
- Department of Dermatology; Jichi Medical University; Shimotsuke Tochigi Japan
| | - Koji Sayama
- Department of Dermatology; University of Ehime; Tou-on-shi Ehime Japan
| | - Kunihiko Tamaki
- Department of Dermatology; University of Tokyo; Bunkyo-ku Tokyo Japan
| | - Mamitaro Ohtsuki
- Department of Dermatology; Jichi Medical University; Shimotsuke Tochigi Japan
| |
Collapse
|
22
|
Rayego-Mateos S, Morgado-Pascual JL, Sanz AB, Ramos AM, Eguchi S, Batlle D, Pato J, Keri G, Egido J, Ortiz A, Ruiz-Ortega M. TWEAK transactivation of the epidermal growth factor receptor mediates renal inflammation. J Pathol 2014; 231:480-94. [PMID: 24037740 DOI: 10.1002/path.4250] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 08/13/2013] [Accepted: 08/21/2013] [Indexed: 11/08/2022]
Abstract
TWEAK, a member of the TNF superfamily, binds to the Fn14 receptor, eliciting biological responses. EGFR signalling is involved in experimental renal injury. Our aim was to investigate the relationship between TWEAK and EGFR in the kidney. Systemic TWEAK administration into C57BL/6 mice increased renal EGFR phosphorylation, mainly in tubular epithelial cells. In vitro, in these cells TWEAK phosphorylated EGFR via Fn14 binding, ADAM17 activation and subsequent release of the EGFR ligands HB-EGF and TGFα. In vivo the EGFR kinase inhibitor Erlotinib inhibited TWEAK-induced renal EGFR activation and downstream signalling, including ERK activation, up-regulation of proinflammatory factors and inflammatory cell infiltration. Moreover, the ADAM17 inhibitor WTACE-2 also prevented those TWEAK-induced renal effects. In vitro TWEAK induction of proinflammatory factors was prevented by EGFR, ERK or ADAM17 inhibition. In contrast, EGFR transactivation did not modify TWEAK-mediated NF-κB activation. Our data suggest that TWEAK transactivates EGFR in the kidney, leading to modulation of downstream effects, including ERK activation and inflammation, and suggest that inhibition of EGFR signalling could be a novel therapeutic tool for renal inflammation.
Collapse
Affiliation(s)
- Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory, Universidad Autónoma, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Hayes BJ, Riehle KJ, Shimizu-Albergine M, Bauer RL, Hudkins KL, Johansson F, Yeh MM, Mahoney WM, Yeung RS, Campbell JS. Activation of platelet-derived growth factor receptor alpha contributes to liver fibrosis. PLoS One 2014; 9:e92925. [PMID: 24667490 PMCID: PMC3965491 DOI: 10.1371/journal.pone.0092925] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 02/27/2014] [Indexed: 01/27/2023] Open
Abstract
Chronic liver injury leads to fibrosis, cirrhosis, and loss of liver function. Liver cirrhosis is the 12th leading cause of death in the United States, and it is the primary risk factor for developing liver cancer. Fibrosis and cirrhosis result from activation of hepatic stellate cells (HSCs), which are the primary collagen producing cell type in the liver. Here, we show that platelet-derived growth factor receptor α (PDGFRα) is expressed by human HSCs, and PDGFRα expression is elevated in human liver disease. Using a green fluorescent protein (GFP) reporter mouse strain, we evaluated the role of PDGFRα in liver disease in mice and found that mouse HSCs express PDGFRα and expression is upregulated during carbon tetrachloride (CCl4) induced liver injury and fibrosis injection. This fibrotic response is reduced in Pdgfrα heterozygous mice, consistent with the hypothesis that liver fibrosis requires upregulation and activation of PDGFRα. These results indicate that Pdgfrα expression is important in the fibrotic response to liver injury in humans and mice, and suggest that blocking PDGFRα–specific signaling pathways in HSCs may provide therapeutic benefit for patients with chronic liver disease.
Collapse
Affiliation(s)
- Brian J. Hayes
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Kimberly J. Riehle
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Department of Surgery, University of Washington, Seattle, Washington, United States of America
| | - Masami Shimizu-Albergine
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Department of Pharmacology, University of Washington, Seattle, Washington, United States of America
| | - Renay L. Bauer
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Kelly L. Hudkins
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Fredrik Johansson
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Matthew M. Yeh
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - William M. Mahoney
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Center for Cardiovascular Biology and Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Raymond S. Yeung
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Department of Surgery, University of Washington, Seattle, Washington, United States of America
| | - Jean S. Campbell
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
24
|
Yamamoto H, Higa-Nakamine S, Noguchi N, Maeda N, Kondo Y, Toku S, Kukita I, Sugahara K. Desensitization by different strategies of epidermal growth factor receptor and ErbB4. J Pharmacol Sci 2014; 124:287-93. [PMID: 24553453 DOI: 10.1254/jphs.13r11cp] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Four transmembrane tyrosine kinases constitute the ErbB protein family: epidermal growth factor receptor (EGFR) or ErbB1, ErbB2, ErbB3, and ErbB4. In general, the structure and mechanism of the activation of these members are similar. However, significant differences in homologous desensitization are known between EGFR and ErbB4. Desensitization of ligand-occupied EGFR occurs by endocytosis, while that of ErbB4 occurs by selective cleavage at the cell surface. Because ErbB4 is abundantly expressed in neurons from fetal to adult brains, elucidation of the desensitization mechanism is important to understand neuronal development and synaptic functions. Recently, it has become clear that heterologous desensitization of EGFR and ErbB4 are induced by endocytosis and cleavage, respectively, similar to homologous desensitization. It has been reported that heterologous desensitization of EGFR is induced by serine phosphorylation of EGFR via the p38 mitogen-activated protein kinase (p38 MAP kinase) pathway in various cell lines, including alveolar epithelial cells. In contrast, the protein kinase C pathway is involved in ErbB4 cleavage. In this review, we will describe recent advances in the desensitization mechanisms of EGFR and ErbB4, mainly in alveolar epithelial cells and hypothalamic neurons, respectively.
Collapse
Affiliation(s)
- Hideyuki Yamamoto
- Department of Biochemistry, Graduate School of Medicine, University of the Ryukyus, Japan
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Tyczewska M, Rucinski M, Ziolkowska A, Szyszka M, Trejter M, Hochol-Molenda A, Nowak KW, Malendowicz LK. Enucleation-induced rat adrenal gland regeneration: expression profile of selected genes involved in control of adrenocortical cell proliferation. Int J Endocrinol 2014; 2014:130359. [PMID: 25431590 PMCID: PMC4241258 DOI: 10.1155/2014/130359] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 07/14/2014] [Accepted: 10/08/2014] [Indexed: 01/01/2023] Open
Abstract
Enucleation-induced adrenal regeneration is a highly controlled process; however, only some elements involved in this process have been recognized. Therefore, we performed studies on regenerating rat adrenals. Microarray RNA analysis and QPCR revealed that enucleation resulted in a rapid elevation of expression of genes involved in response to wounding, defense response, and in immunological processes. Factors encoded by these genes obscure possible priming effects of various cytokines on initiation of regeneration. In regenerating adrenals we identified over 100 up- or downregulated genes involved in adrenocortical cell proliferation. The changes were most significant at days 2-3 after enucleation and their number decreased during regeneration. For example, expression analysis revealed a notable upregulation of the growth arrest gene, Gadd45, only 24 hours after surgery while expression of cyclin B1 and Cdk1 genes was notably elevated between days 1-8 of regeneration. These changes were accompanied by changes in expression levels of numerous growth factors and immediate-early transcription factors genes. Despite notable differences in mechanisms of adrenal and liver regeneration, in regenerating adrenals we identified genes, the expression of which is well recognized in regenerating liver. Thus, it seems legitimate to suggest that, in the rat, the general model of liver and adrenal regeneration demonstrate some degree of similarity.
Collapse
Affiliation(s)
- Marianna Tyczewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marcin Rucinski
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Agnieszka Ziolkowska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marta Szyszka
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marcin Trejter
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Anna Hochol-Molenda
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Krzysztof W. Nowak
- Department of Animal Physiology and Biochemistry, Poznan University of Life Sciences, Poznan, Poland
| | - Ludwik K. Malendowicz
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
- *Ludwik K. Malendowicz:
| |
Collapse
|
26
|
Abstract
Liver regeneration is perhaps the most studied example of compensatory growth aimed to replace loss of tissue in an organ. Hepatocytes, the main functional cells of the liver, manage to proliferate to restore mass and to simultaneously deliver all functions hepatic functions necessary to maintain body homeostasis. They are the first cells to respond to regenerative stimuli triggered by mitogenic growth factor receptors MET (the hepatocyte growth factor receptor] and epidermal growth factor receptor and complemented by auxiliary mitogenic signals induced by other cytokines. Termination of liver regeneration is a complex process affected by integrin mediated signaling and it restores the organ to its original mass as determined by the needs of the body (hepatostat function). When hepatocytes cannot proliferate, progenitor cells derived from the biliary epithelium transdifferentiate to restore the hepatocyte compartment. In a reverse situation, hepatocytes can also transdifferentiate to restore the biliary compartment. Several hormones and xenobiotics alter the hepatostat directly and induce an increase in liver to body weight ratio (augmentative hepatomegaly). The complex challenges of the liver toward body homeostasis are thus always preserved by complex but unfailing responses involving orchestrated signaling and affecting growth and differentiation of all hepatic cell types.
Collapse
Affiliation(s)
- George K Michalopoulos
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
27
|
Liu FL, Wu CC, Chang DM. TACE-dependent amphiregulin release is induced by IL-1β and promotes cell invasion in fibroblast-like synoviocytes in rheumatoid arthritis. Rheumatology (Oxford) 2013; 53:260-9. [PMID: 24196392 DOI: 10.1093/rheumatology/ket350] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES The aims of this study were to investigate the expression of amphiregulin (AREG) and TNF-α-converting enzyme (TACE) in fibroblast-like synoviocytes from humans with RA (FLS-RA) when stimulated with proinflammatory cytokines and to explore whether AREG plays a role in RA. METHODS The effects of cytokines on the expression of AREG and TACE in FLS-RA were measured by quantitative RT-PCR and western blotting. Blockade of IL-1β-mediated pathways was used to verify the involvement of intracellular signal pathways in the induction of AREG and TACE. TAPI-1 and TACE short hairpin RNA (shRNA) infection were used to identify the role of TACE in IL-1β-induced AREG secretion and shedding. AREG-induced production of MMP-1 and cadherin-11 in FLS-RA were measured by ELISA or western blotting. The effect of AREG on FLS-RA invasion was examined using a Transwell invasion assay. RESULTS IL-1β, but not other tested cytokines, increased the expressions of AREG mRNA and protein in a dose-responsive and time-dependent manner in FLS-RA. IL-1β induced AREG expression via p38 MAPK, NF-κB, JNK and ERK1/2 signalling pathways and induced TACE expression via PI3K, p38MAPK and NF-κB signalling pathways in FLS-RA. TACE mediated AREG secretion and shedding. EGFR (ErbB1) and Her-2 (ErbB2) were expressed in FLS-RA, and AREG increased MMP-1 and cadherin-11 expression in FLS-RA. AREG promoted the FLS-RA invasion ability. CONCLUSION AREG and TACE expression were up-regulated by IL-1β and their activations on FLS-RA lead to the matrix degradation by inducing MMP-1 and cadherin-11 production. TACE activity is necessary for IL-1β-induced AREG release. Our results demonstrate that IL-1β-induced AREG release may be involved in the pathogenesis of RA.
Collapse
Affiliation(s)
- Fei-Lan Liu
- Rheumatology/Immunology/Allergy, Tri-Service General Hospital, 325 Cheng-Kung Road, Section 2, Neihu 114, Taipei, Taiwan, Republic of China.
| | | | | |
Collapse
|
28
|
McMahan RS, Riehle KJ, Fausto N, Campbell JS. A disintegrin and metalloproteinase 17 regulates TNF and TNFR1 levels in inflammation and liver regeneration in mice. Am J Physiol Gastrointest Liver Physiol 2013; 305:G25-34. [PMID: 23639813 PMCID: PMC3725689 DOI: 10.1152/ajpgi.00326.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
A disintegrin and metalloproteinase 17 (ADAM17), or tumor necrosis factor (TNF)-α-converting enzyme, is a key metalloproteinase and physiological convertase for a number of putative targets that play critical roles in cytokine and growth factor signaling. These interdependent pathways are essential components of the signaling network that links liver function with the compensatory growth that occurs during liver regeneration following 2/3 partial hepatectomy (PH) or chemically induced hepatotoxicity. Despite identification of many soluble factors needed for efficient liver regeneration, very little is known about how such ligands are regulated in the liver. To directly study the role of ADAM17 in the liver, we employed two cell-specific ADAM17 knockout (KO) mouse models. Using lipopolysaccharide (LPS) as a robust stimulus for TNF release, we found attenuated levels of circulating TNF in myeloid-specific ADAM17 KO mice (ADAM17 m-KO) and, unexpectedly, in mice with hepatocyte-specific ADAM17 deletion (ADAM17 h-KO), indicating that ADAM17 expression in both cell types plays a role in TNF shedding. After 2/3 PH, induction of TNF, TNFR1, and amphiregulin (AR) was significantly attenuated in ADAM17 h-KO mice, implicating ADAM17 as the primary sheddase for these factors in the liver. Surprisingly, the extent and timing of hepatocyte proliferation were not affected after PH or carbon tetrachloride injection in ADAM17 h-KO or ADAM17 m-KO mice. We conclude that ADAM17 regulates TNF, TNFR1, and AR in the liver, and its expression in both hepatocytes and myeloid cells is important for TNF regulation after LPS injury or 2/3 PH, but is not required for liver regeneration.
Collapse
Affiliation(s)
- Ryan S. McMahan
- 1Department of Pathology, University of Washington, Seattle, Washington; and
| | - Kimberly J. Riehle
- 1Department of Pathology, University of Washington, Seattle, Washington; and ,2Department of Surgery, University of Washington, Seattle, Washington
| | - Nelson Fausto
- 1Department of Pathology, University of Washington, Seattle, Washington; and
| | - Jean S. Campbell
- 1Department of Pathology, University of Washington, Seattle, Washington; and
| |
Collapse
|
29
|
Waheed F, Dan Q, Amoozadeh Y, Zhang Y, Tanimura S, Speight P, Kapus A, Szászi K. Central role of the exchange factor GEF-H1 in TNF-α-induced sequential activation of Rac, ADAM17/TACE, and RhoA in tubular epithelial cells. Mol Biol Cell 2013; 24:1068-82. [PMID: 23389627 PMCID: PMC3608494 DOI: 10.1091/mbc.e12-09-0661] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Tumor necrosis factor-α activates the enzyme TACE/ADAM17 through the guanine nucleotide exchange factor GEF-H1, Rac, and p38, leading to activation of the epidermal growth factor. GEF-H1 mediates hierarchical activation of Rac and RhoA through differential phosphorylation. Transactivation of the epidermal growth factor receptor (EGFR) by tumor necrosis factor-α (TNF-α) is a key step in mediating RhoA activation and cytoskeleton and junction remodeling in the tubular epithelium. In this study we explore the mechanisms underlying TNF-α–induced EGFR activation. We show that TNF-α stimulates the TNF-α convertase enzyme (TACE/a disintegrin and metalloproteinase-17), leading to activation of the EGFR/ERK pathway. TACE activation requires the mitogen-activated protein kinase p38, which is activated through the small GTPase Rac. TNF-α stimulates both Rac and RhoA through the guanine nucleotide exchange factor (GEF)-H1 but by different mechanisms. EGFR- and ERK-dependent phosphorylation at the T678 site of GEF-H1 is a prerequisite for RhoA activation only, whereas both Rac and RhoA activation require GEF-H1 phosphorylation on S885. Of interest, GEF-H1-mediated Rac activation is upstream from the TACE/EGFR/ERK pathway and regulates T678 phosphorylation. We also show that TNF-α enhances epithelial wound healing through TACE, ERK, and GEF-H1. Taken together, our findings can explain the mechanisms leading to hierarchical activation of Rac and RhoA by TNF-α through a single GEF. This mechanism could coordinate GEF functions and fine-tune Rac and RhoA activation in epithelial cells, thereby promoting complex functions such as sheet migration.
Collapse
Affiliation(s)
- Faiza Waheed
- Department of Surgery, University of Toronto, Toronto, ON M5B 1T8, Canada
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Phosphorylation of epidermal growth factor receptor at serine 1047 by MAP kinase-activated protein kinase-2 in cultured lung epithelial cells treated with flagellin. Arch Biochem Biophys 2013; 529:75-85. [DOI: 10.1016/j.abb.2012.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 11/16/2012] [Accepted: 11/17/2012] [Indexed: 11/23/2022]
|
31
|
Corlu A, Loyer P. Regulation of the g1/s transition in hepatocytes: involvement of the cyclin-dependent kinase cdk1 in the DNA replication. Int J Hepatol 2012; 2012:689324. [PMID: 23091735 PMCID: PMC3471441 DOI: 10.1155/2012/689324] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 08/29/2012] [Indexed: 12/16/2022] Open
Abstract
A singular feature of adult differentiated hepatocytes is their capacity to proliferate allowing liver regeneration. This review emphasizes the literature published over the last 20 years that established the most important pathways regulating the hepatocyte cell cycle. Our article also aimed at illustrating that many discoveries in this field benefited from the combined use of in vivo models of liver regeneration and in vitro models of primary cultures of human and rodent hepatocytes. Using these models, our laboratory has contributed to decipher the different steps of the progression into the G1 phase and the commitment to S phase of proliferating hepatocytes. We identified the mitogen dependent restriction point located at the two-thirds of the G1 phase and the concomitant expression and activation of both Cdk1 and Cdk2 at the G1/S transition. Furthermore, we demonstrated that these two Cdks contribute to the DNA replication. Finally, we provided strong evidences that Cdk1 expression and activation is correlated to extracellular matrix degradation upon stimulation by the pro-inflammatory cytokine TNFα leading to the identification of a new signaling pathway regulating Cdk1 expression at the G1/S transition. It also further confirms the well-orchestrated regulation of liver regeneration via multiple extracellular signals and pathways.
Collapse
Affiliation(s)
- Anne Corlu
- Inserm UMR S 991, Foie Métabolismes et Cancer, Université de Rennes 1, Hôpital Pontchaillou, 35033 Rennes Cedex, France
| | - Pascal Loyer
- Inserm UMR S 991, Foie Métabolismes et Cancer, Université de Rennes 1, Hôpital Pontchaillou, 35033 Rennes Cedex, France
| |
Collapse
|
32
|
The EGF receptor and HER2 participate in TNF-α-dependent MAPK activation and IL-8 secretion in intestinal epithelial cells. Mediators Inflamm 2012; 2012:207398. [PMID: 22988345 PMCID: PMC3440955 DOI: 10.1155/2012/207398] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Accepted: 07/24/2012] [Indexed: 12/29/2022] Open
Abstract
TNF-α activates multiple mitogen-activated protein kinase (MAPK) cascades in intestinal epithelial cells (IECs) leading to the secretion of interleukin 8 (IL-8), a neutrophil chemoattractant and an angiogenic factor with tumor promoting properties. As the epidermal growth factor receptor (EGFR) is a known transducer of proliferative signals and a potent activator of MAPKs, we hypothesized that the EGFR participates in TNF-dependent MAPK activation and IL-8 secretion by intestinal epithelial cells (IECs).
We show that the EGFR is tyrosine-phosphorylated following treatment of IECs (HT-29 and IEC-6) with TNF-α. This requires EGFR autophosphorylation as it was blocked by the EGFR kinase inhibitor AG1478. Autophosphorylation was also inhibited by both a Src-kinase inhibitor and the metalloproteinase inhibitor batimastat. TNF treatment of IECs resulted in the accumulation of soluble TGF-α; treatment of IECs with batimastat suppressed TGF-α release and immunoneutralization of TGF-α resulted in decreased EGFR and ERK phosphorylations. TNF-α treatment of IECs resulted in an association between EGFR and HER2 and inhibition of HER2 using a specific inhibitor AG879 in combination with AG1478-suppressed TNF-α-dependent ERK phosphorylation and IL-8 release. Downregulation of HER2 via siRNA resulted in a significant decrease in ERK phosphorylation and a 50% reduction in IL-8 secretion.
Collapse
|
33
|
Taki-Eldin A, Zhou L, Xie HY, Zheng SS. Liver regeneration after liver transplantation. ACTA ACUST UNITED AC 2012; 48:139-53. [PMID: 22572792 DOI: 10.1159/000337865] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 02/07/2012] [Indexed: 12/14/2022]
Abstract
BACKGROUND/PURPOSE The liver has a remarkable capacity to regenerate after injury or resection. The aim of this review is to outline the mechanisms and factors affecting liver regeneration after liver transplantation. METHODS Relevant studies were reviewed using Medline, PubMed and Springer databases. RESULTS A variety of cytokines (such as interleukin-6 and tumor necrosis factor-α), growth factors (like hepatocyte growth factor and transforming growth factor-α) and cells are involved in liver regeneration. Several factors affect liver regeneration after transplantation such as ischemic injury, graft size, immunosuppression, steatosis, donor age and viral hepatitis. CONCLUSION Liver regeneration has been studied for many years. However, further research is essential to reveal the complex processes affecting liver regeneration, which may provide novel strategies in the management of liver transplantation recipients and donors.
Collapse
Affiliation(s)
- A Taki-Eldin
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | | | | |
Collapse
|
34
|
EGFR: A Master Piece in G1/S Phase Transition of Liver Regeneration. Int J Hepatol 2012; 2012:476910. [PMID: 23050157 PMCID: PMC3461622 DOI: 10.1155/2012/476910] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 07/11/2012] [Indexed: 02/07/2023] Open
Abstract
Unraveling the molecular clues of liver proliferation has become conceivable thanks to the model of two-third hepatectomy. The synchronicity and the well-scheduled aspect of this process allow scientists to slowly decipher this mystery. During this phenomenon, quiescent hepatocytes of the remnant lobes are able to reenter into the cell cycle initiating the G1-S progression synchronously before completing the cell cycle. The major role played by this step of the cell cycle has been emphasized by loss-of-function studies showing a delay or a lack of coordination in the hepatocytes G1-S progression. Two growth factor receptors, c-Met and EGFR, tightly drive this transition. Due to the level of complexity surrounding EGFR signaling, involving numerous ligands, highly controlled regulations and multiple downstream pathways, we chose to focus on the EGFR pathway for this paper. We will first describe the EGFR pathway in its integrity and then address its essential role in the G1/S phase transition for hepatocyte proliferation. Recently, other levels of control have been discovered to monitor this pathway, which will lead us to discuss regulations of the EGFR pathway and highlight the potential effect of misregulations in pathologies.
Collapse
|
35
|
Maillé E, Trinh NTN, Privé A, Bilodeau C, Bissonnette É, Grandvaux N, Brochiero E. Regulation of normal and cystic fibrosis airway epithelial repair processes by TNF-α after injury. Am J Physiol Lung Cell Mol Physiol 2011; 301:L945-55. [DOI: 10.1152/ajplung.00149.2011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic infection and inflammation have been associated with progressive airway epithelial damage in patients with cystic fibrosis (CF). However, the effect of inflammatory products on the repair capacity of respiratory epithelia is unclear. Our objective was to study the regulation of repair mechanisms by tumor necrosis factor-α (TNF-α), a major component of inflammation in CF, in a model of mechanical wounding, in two bronchial cell lines, non-CF NuLi and CF CuFi. We observed that TNF-α enhanced the NuLi and CuFi repair rates. Chronic exposure (24–48 h) to TNF-α augmented this stimulation as well as the migration rate during repair. The cellular mechanisms involved in this stimulation were then evaluated. First, we discerned that TNF-α induced metalloproteinase-9 release, epidermal growth factor (EGF) shedding, and subsequent EGF receptor transactivation. Second, TNF-α-induced stimulation of the NuLi and CuFi wound-closure rates was prevented by GM6001 (metalloproteinase inhibitor), EGF antibody (to titrate secreted EGF), and EGF receptor tyrosine kinase inhibitors. Furthermore, we recently reported a relationship between the EGF response and K+channel function, both controlling bronchial repair. We now show that TNF-α enhances KvLQT1 and KATPcurrents, while their inhibition abolishes TNF-α-induced repair stimulation. These results indicate that the effect of TNF-α is mediated, at least in part, through EGF receptor transactivation and K+channel stimulation. In contrast, cell proliferation during repair was slowed by TNF-α, suggesting that TNF-α could exert contrasting actions on repair mechanisms of CF airway epithelia. Finally, the stimulatory effect of TNF-α on airway wound repair was confirmed on primary airway epithelial cells, from non-CF and CF patients.
Collapse
Affiliation(s)
- Emilie Maillé
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM)—Hôtel-Dieu, Montréal
- Département de médecine and
| | - Nguyen Thu Ngan Trinh
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM)—Hôtel-Dieu, Montréal
- Département de médecine and
| | - Anik Privé
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM)—Hôtel-Dieu, Montréal
| | - Claudia Bilodeau
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM)—Hôtel-Dieu, Montréal
| | - Élyse Bissonnette
- Institut Universitaire de cardiologie et de pneumologie de Québec, Département de médecine, Université Laval, Québec, Québec, Canada
| | - Nathalie Grandvaux
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM)—Hôtel-Dieu, Montréal
- Département de Biochimie, Université de Montréal, Montréal; and
| | - Emmanuelle Brochiero
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM)—Hôtel-Dieu, Montréal
- Département de médecine and
| |
Collapse
|
36
|
Hobbs SS, Goettel JA, Liang D, Yan F, Edelblum KL, Frey MR, Mullane MT, Polk DB. TNF transactivation of EGFR stimulates cytoprotective COX-2 expression in gastrointestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2011; 301:G220-9. [PMID: 21566012 PMCID: PMC3154604 DOI: 10.1152/ajpgi.00383.2010] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
TNF and epidermal growth factor (EGF) are well-known stimuli of cyclooxygenase (COX)-2 expression, and TNF stimulates transactivation of EGF receptor (EGFR) signaling to promote survival in colon epithelial cells. We hypothesized that COX-2 induction and cell survival signaling downstream of TNF are mediated by EGFR transactivation. TNF treatment was more cytotoxic to COX-2(-/-) mouse colon epithelial (MCE) cells than wild-type (WT) young adult mouse colon (YAMC) epithelial cells or COX-1(-/-) cells. TNF also induced COX-2 protein and mRNA expression in YAMC cells, but blockade of EGFR kinase activity or expression inhibited COX-2 upregulation. TNF-induced COX-2 expression was reduced and absent in EGFR(-/-) and TNF receptor-1 (TNFR1) knockout MCE cells, respectively, but was restored upon expression of the WT receptors. Inhibition of mediators of EGFR transactivation, Src family kinases and p38 MAPK, blocked TNF-induced COX-2 protein and mRNA expression. Finally, TNF injection increased COX-2 expression in colon epithelium of WT, but not kinase-defective EGFR(wa2) and EGFR(wa5), mice. These data indicate that TNFR1-dependent transactivation of EGFR through a p38- and/or an Src-dependent mechanism stimulates COX-2 expression to promote cell survival. This highlights an EGFR-dependent cell signaling pathway and response that may be significant in colitis-associated carcinoma.
Collapse
Affiliation(s)
- Stuart S. Hobbs
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics,
| | - Jeremy A. Goettel
- 2Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Dongchun Liang
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics,
| | - Fang Yan
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics,
| | - Karen L. Edelblum
- 2Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Mark R. Frey
- 3Departments of Pediatrics and Biochemistry and Molecular Biology, The Saban Research Institute of Children's Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles, California
| | - Matthew T. Mullane
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics,
| | - D. Brent Polk
- 3Departments of Pediatrics and Biochemistry and Molecular Biology, The Saban Research Institute of Children's Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles, California
| |
Collapse
|
37
|
Kakiashvili E, Dan Q, Vandermeer M, Zhang Y, Waheed F, Pham M, Szászi K. The epidermal growth factor receptor mediates tumor necrosis factor-alpha-induced activation of the ERK/GEF-H1/RhoA pathway in tubular epithelium. J Biol Chem 2011; 286:9268-79. [PMID: 21212278 PMCID: PMC3059019 DOI: 10.1074/jbc.m110.179903] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2010] [Revised: 01/05/2011] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor (TNF)-α induces cytoskeleton and intercellular junction remodeling in tubular epithelial cells; the underlying mechanisms, however, are incompletely explored. We have previously shown that ERK-mediated stimulation of the RhoA GDP/GTP exchange factor GEF-H1/Lfc is critical for TNF-α-induced RhoA stimulation. Here we investigated the upstream mechanisms of ERK/GEF-H1 activation. Surprisingly, TNF-α-induced ERK and RhoA stimulation in tubular cells were prevented by epidermal growth factor receptor (EGFR) inhibition or silencing. TNF-α also enhanced phosphorylation of the EGFR. EGF treatment mimicked the effects of TNF-α, as it elicited potent, ERK-dependent GEF-H1 and RhoA activation. Moreover, EGF-induced RhoA activation was prevented by GEF-H1 silencing, indicating that GEF-H1 is a key downstream effector of the EGFR. The TNF-α-elicited EGFR, ERK, and RhoA stimulation were mediated by the TNF-α convertase enzyme (TACE) that can release EGFR ligands. Further, EGFR transactivation also required the tyrosine kinase Src, as Src inhibition prevented TNF-α-induced activation of the EGFR/ERK/GEF-H1/RhoA pathway. Importantly, a bromodeoxyuridine (BrdU) incorporation assay and electric cell substrate impedance-sensing (ECIS) measurements revealed that TNF-α stimulated cell growth in an EGFR-dependent manner. In contrast, TNF-α-induced NFκB activation was not prevented by EGFR or Src inhibition, suggesting that TNF-α exerts both EGFR-dependent and -independent effects. In summary, in the present study we show that the TNF-α-induced activation of the ERK/GEF-H1/RhoA pathway in tubular cells is mediated through Src- and TACE-dependent EGFR activation. Such a mechanism could couple inflammatory and proliferative stimuli and, thus, may play a key role in the regulation of wound healing and fibrogenesis.
Collapse
Affiliation(s)
- Eli Kakiashvili
- From the Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Department of Surgery, University of Toronto, Toronto, Ontario M5B 1W8, Canada
| | - Qinghong Dan
- From the Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Department of Surgery, University of Toronto, Toronto, Ontario M5B 1W8, Canada
| | - Matthew Vandermeer
- From the Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Department of Surgery, University of Toronto, Toronto, Ontario M5B 1W8, Canada
| | - Yuqian Zhang
- From the Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Department of Surgery, University of Toronto, Toronto, Ontario M5B 1W8, Canada
| | - Faiza Waheed
- From the Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Department of Surgery, University of Toronto, Toronto, Ontario M5B 1W8, Canada
| | - Monica Pham
- From the Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Department of Surgery, University of Toronto, Toronto, Ontario M5B 1W8, Canada
| | - Katalin Szászi
- From the Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Department of Surgery, University of Toronto, Toronto, Ontario M5B 1W8, Canada
| |
Collapse
|
38
|
Hora C, Romanque P, Dufour JFF. Effect of sorafenib on murine liver regeneration. Hepatology 2011; 53:577-86. [PMID: 21274878 DOI: 10.1002/hep.24037] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2010] [Accepted: 09/28/2010] [Indexed: 02/05/2023]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) is a common cause of cancer-related death. Sorafenib prolongs survival of patients with advanced disease and is approved for the systemic treatment of unresectable HCC. It possesses antiangiogenic and antiproliferative properties by way of inhibition of the receptor tyrosine kinases vascular endothelial growth factor receptor 2 (VEGFR-2) and platelet-derived growth factor receptor-beta 1/2 (PDGFR-β) and the kinase RAF. Sorafenib represents a candidate compound for adjuvant therapy in HCC patients. The aim of our study was to investigate whether sorafenib affects liver regeneration. C57BL6 mice received sorafenib orally at 30 mg/kg/day or its vehicle either for 14 days until the day before hepatectomy or starting the day after surgery or both. Animals were sacrificed 24, 72, and 120 hours after hepatectomy. Liver regeneration was calculated as a percent of initial liver weight. Bromodeoxyuridine (BrdU) incorporation and phospho-extracellular signal-regulated kinase (pERK1/2) were determined by immunohistochemistry on liver sections. VEGF-A, PDGF-BB, and hepatocyte growth factor (HGF) levels were measured in liver tissue homogenates. Histological analysis of scar tissue was performed. Treatment stopped 1 day before surgery had no impact on liver regeneration. Continuous sorafenib treatment and treatment started 1 day after surgery had statistically significant effects on liver regeneration at 120 hours compared to vehicle-treated control animals (72% ± 12 versus control 88% ± 15 and 70% ± 13 versus control 86% ± 5 at 120 hours, both P ≤ 0.02). BrdU incorporation showed decreased numbers of positive nuclei in both groups receiving sorafenib after surgery. Phospho-ERK levels were reduced in sorafenib-treated animals. An increase of VEGF-A levels was observed in mice receiving sorafenib. Wound-healing complications were observed in animals receiving sorafenib after surgery and confirmed on histological sections. CONCLUSION This preclinical study shows that sorafenib did not impact on liver regeneration when ceased before surgery; however, administration after hepatectomy affected late liver regeneration.
Collapse
Affiliation(s)
- Caroline Hora
- Department of Clinical Research, University of Bern, Switzerland
| | | | | |
Collapse
|
39
|
|
40
|
Atta HM. Gene therapy for liver regeneration: experimental studies and prospects for clinical trials. World J Gastroenterol 2010; 16:4019-30. [PMID: 20731015 PMCID: PMC2928455 DOI: 10.3748/wjg.v16.i32.4019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Revised: 03/03/2010] [Accepted: 03/10/2010] [Indexed: 02/06/2023] Open
Abstract
The liver is an exceptional organ, not only because of its unique anatomical and physiological characteristics, but also because of its unlimited regenerative capacity. Unfolding of the molecular mechanisms that govern liver regeneration has allowed researchers to exploit them to augment liver regeneration. Dramatic progress in the field, however, was made by the introduction of the powerful tool of gene therapy. Transfer of genetic materials, such as hepatocyte growth factor, using both viral and non-viral vectors has proved to be successful in augmenting liver regeneration in various animal models. For future clinical studies, ongoing research aims at eliminating toxicity of viral vectors and increasing transduction efficiency of non-viral vectors, which are the main drawbacks of these systems. Another goal of current research is to develop gene therapy that targets specific liver cells using receptors that are unique to and highly expressed by different liver cell types. The outcome of such investigations will, undoubtedly, pave the way for future successful clinical trials.
Collapse
|
41
|
Campbell JS, Argast GM, Yuen SY, Hayes B, Fausto N. Inactivation of p38 MAPK during liver regeneration. Int J Biochem Cell Biol 2010; 43:180-8. [PMID: 20708092 DOI: 10.1016/j.biocel.2010.08.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 07/27/2010] [Accepted: 08/03/2010] [Indexed: 12/22/2022]
Abstract
There is increasing evidence that p38 MAPK, which is classified as a stress-activated kinase, also participates in cell cycle regulation, functioning as a suppressor of cell proliferation and tumorigenesis. We conducted a study of p38 MAPK phosphorylation during liver regeneration in mice to determine whether p38 MAPK activation or inactivation may correlate with events that lead to DNA replication after partial hepatectomy (PH), and whether p38 MAPK activation may be required for hepatocyte DNA replication in vivo and in culture. We report that active p38 (Pi-p38 MAPK) is present in normal liver, is rapidly inactivated starting 30 min after PH, and is re-activated by 12h. Although levels of Pi-MKK 3/6, the upstream kinases that activate p38 MAPK increase after PH, the expression of the dual protein phosphatase 1 is also elevated, and may be responsible for Pi-p38 MAPK dephosphorylation after PH. Inactivation and re-activation of p38 MAPK inversely correlates with the stimulation of protein synthesis and translation pathways, as indicated by activation of p70S6 kinase, increases in the phosphorylation of initiation factor elF-4E and translational repressor, 4E-BP. The activity of a p38 MAPK downstream substrate, MAPKAPK2 (MK2), did not reflect the changing levels of Pi-p38 MAPK during liver regeneration. Pi-p38 MAPK may be involved in TNF-stimulated DNA replication of murine hepatocytes in culture, but is not necessary for hepatocyte DNA replication after PH. Our results suggest that p38 MAPK inactivation plays a permissible role in DNA replication during liver regeneration and is consistent with a role for p38 MAPK in the maintenance of hepatocyte cell cycle arrest in adult liver.
Collapse
Affiliation(s)
- Jean S Campbell
- Department of Pathology, University of Washington, School of Medicine, Seattle, WA 98195, USA.
| | | | | | | | | |
Collapse
|
42
|
Park YJ, Lee H, Lee JH. Macrophage inhibitory cytokine-1 transactivates ErbB family receptors via the activation of Src in SK-BR-3 human breast cancer cells. BMB Rep 2010; 43:91-6. [PMID: 20193126 DOI: 10.5483/bmbrep.2010.43.2.091] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The function of macrophage inhibitory cytokine-1 (MIC-1) in cancer remains controversial, and its signaling pathways remain poorly understood. In this study, we demonstrate that MIC-1 induces the transactivation of EGFR, ErbB2, and ErbB3 through the activation of c-Src in SK-BR-3 breast cells. MIC-1 induced significant phosphorylation of EGFR at Tyr845, ErbB2 at Tyr877, and ErbB3 at Tyr1289 as well as Akt and p38, Erk1/2, and JNK mitogen-activated protein kinases (MAPKs). Treatment of SK-BR-3 cells with MIC-1 increased the phosphorylation level of Src at Tyr416, and induced invasiveness of those cells. Inhibition of c-Src activity resulted in the complete abolition of MIC-1-induced phosphorylation of the EGFR, ErbB2, and ErbB3, as well as invasiveness and matrix metalloproteinase (MMP)-9 expression in SK-BR-3 cells. Collectively, these results show that MIC-1 may participate in the malignant progression of certain cancer cells through the activation of c-Src, which in turn may transactivate ErbB-family receptors.
Collapse
Affiliation(s)
- Yun Jung Park
- Departments of Biochemistry and College of Natural Sciences, Kangwon National University, Chuncheon 200-701, Korea
| | | | | |
Collapse
|
43
|
Zhang F, Shu R, Wu X, Zhao X, Feng D, Wang L, Lu S, Liu Q, Xiang Y, Fei J, Huang L, Wang Z. Delayed liver injury and impaired hepatocyte proliferation after carbon tetrachloride exposure in BPOZ2-deficient mice. Toxicol Lett 2009; 188:201-7. [PMID: 19393728 DOI: 10.1016/j.toxlet.2009.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 04/11/2009] [Accepted: 04/14/2009] [Indexed: 12/28/2022]
Abstract
BPOZ2 is a tumor suppressive mediator in PTEN signaling pathway and plays an important role in cell proliferation. In this study, we investigated the physiology functions of BPOZ2 in CCl(4)-induced liver injury and hepatocyte proliferation afterwards. After acute CCl(4) administration, BPOZ2 null mice exhibited delayed liver injury and impaired hepatocyte proliferation, which was accompanied by altered kinetics of CYP2E1 protein expression, compromised cyclin D1 expression and shortened duration of ERK activation. These results for the first time define that BPOZ2 is an important regulator involved in the injury and repair process induced by acute CC1(4) administration in mouse liver.
Collapse
Affiliation(s)
- Feng Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Berasain C, Perugorria MJ, Latasa MU, Castillo J, Goñi S, Santamaría M, Prieto J, Avila MA. The epidermal growth factor receptor: a link between inflammation and liver cancer. Exp Biol Med (Maywood) 2009; 234:713-25. [PMID: 19429859 DOI: 10.3181/0901-mr-12] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Epidemiological studies have established that many tumours occur in association with persistent inflammation. One clear example of inflammation-related cancer is hepatocellular carcinoma (HCC). HCC slowly unfolds on a background of chronic inflammation triggered by exposure to infectious agents (hepatotropic viruses), toxic compounds (ethanol), or metabolic impairment. The molecular links that connect inflammation and cancer are not completely known, but evidence gathered over the past few years is beginning to define the precise mechanisms. A central role for cytokines such as interleukin-6 (IL-6) and IL-1 (alpha and beta) in liver cancer has been established in experimental models. Besides these inflammatory mediators, mounting evidence points to the dysregulation of specific growth and survival-related pathways in HCC development. Among them is the pathway governed by the epidermal growth factor receptor (EGFR), which can be bound and activated by a broad family of ligands. Of special relevance is the fact that the EGFR engages in extensive crosstalk with other signaling pathways, serving as a "signaling hub" for an increasing list of growth factors, cytokines, and inflammatory mediators. In this review, we summarize the most recent evidences supporting a role for the EGFR system in inflammation-related cell signaling, with special emphasis in liver inflammation and HCC. The molecular dissection of the pathways connecting the inflammatory reaction and neoplasia will facilitate the development of novel and more effective antitumor strategies.
Collapse
Affiliation(s)
- Carmen Berasain
- Division of Hepatology and Gene Therapy, CIMA-Universidad de Navarra, 31008 Pamplona, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Shin MS, Shinghirunnusorn P, Sugishima Y, Nishimura M, Suzuki S, Koizumi K, Saiki I, Sakurai H. Cross interference with TNF-alpha-induced TAK1 activation via EGFR-mediated p38 phosphorylation of TAK1-binding protein 1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:1156-64. [PMID: 19393267 DOI: 10.1016/j.bbamcr.2009.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2008] [Revised: 03/31/2009] [Accepted: 04/13/2009] [Indexed: 12/17/2022]
Abstract
Transforming growth factor-alpha-activated kinase 1 (TAK1) has been widely recognized as a kinase that regulates multiple intracellular signaling pathways evoked by cytokines and immune receptor activation. We have recently reported that tumor necrosis factor-alpha (TNF-alpha) triggers internalization of epidermal growth factor receptor (EGFR) through a TAK1-p38alpha signaling pathway, which results in a transient suppression of the EGFR. In the present study, we investigated the pathway of intracellular signaling in the opposite direction. Ligand-induced activation of EGFR caused phosphorylation of the TAK1-binding proteins TAB1 and TAB2 in a TAK1-independent manner. EGFR-mediated phosphorylation of TAB1 was completely inhibited by a chemical inhibitor and siRNA of p38alpha. The phosphorylation of TAB1 was occurred at Ser-423 and Thr-431, the residues underlying the p38-mediated feedback inhibition of TAK1. In contrast, phosphorylation of TAB2 was sustained, and largely resistant to p38 inhibition. The inducible phosphorylation of TAB1 interfered with a response of EGF-treated cells to TNF-alpha-induced TAK1 activation, which led to the reduction of NF-kappaB activation. Collectively, these results demonstrated that EGFR activation interfered with TNF-alpha-induced TAK1 activation via p38-mediated phosphorylation of TAB1.
Collapse
Affiliation(s)
- Myoung-Sook Shin
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Smith PC, Guerrero J, Tobar N, Cáceres M, González MJ, Martínez J. Tumor necrosis factor-α-stimulated membrane type 1-matrix metalloproteinase production is modulated by epidermal growth factor receptor signaling in human gingival fibroblasts. J Periodontal Res 2009; 44:73-80. [DOI: 10.1111/j.1600-0765.2007.01081.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
47
|
Shepard HM, Brdlik CM, Schreiber H. Signal integration: a framework for understanding the efficacy of therapeutics targeting the human EGFR family. J Clin Invest 2009; 118:3574-81. [PMID: 18982164 DOI: 10.1172/jci36049] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The human EGFR (HER) family is essential for communication between many epithelial cancer cell types and the tumor microenvironment. Therapeutics targeting the HER family have demonstrated clinical success in the treatment of diverse epithelial cancers. Here we propose that the success of HER family-targeted monoclonal antibodies in cancer results from their ability to interfere with HER family consolidation of signals initiated by a multitude of other receptor systems. Ligand/receptor systems that initiate these signals include cytokine receptors, chemokine receptors, TLRs, GPCRs, and integrins. We further extrapolate that improvements in cancer therapeutics targeting the HER family are likely to incorporate mechanisms that block or reverse stromal support of malignant progression by isolating the HER family from autocrine and stromal influences.
Collapse
|
48
|
Frey MR, Edelblum KL, Mullane MT, Liang D, Polk DB. The ErbB4 growth factor receptor is required for colon epithelial cell survival in the presence of TNF. Gastroenterology 2009; 136:217-26. [PMID: 18973758 PMCID: PMC2811086 DOI: 10.1053/j.gastro.2008.09.023] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Revised: 08/12/2008] [Accepted: 09/18/2008] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS The ErbB4 receptor tyrosine kinase regulates cell growth, survival, and differentiation in several tissues, but its role in the gastrointestinal tract has not been reported. We tested the hypothesis that ErbB4 promotes intestinal cell survival and restitution following injury or inflammation. METHODS ErbB4 expression in human inflammatory bowel disease was determined by immunohistochemistry. Mice were subjected to dextran sulfate sodium (DSS, 3%) colitis or injected with tumor necrosis factor (TNF), and ErbB4 expression was quantified by immunohistochemistry and Western blot. Cultured young adult mouse colon (YAMC) cells were exposed to TNF, and ErbB4 messenger RNA, protein, and phosphorylation levels were measured. Cells transfected with ErbB4 small interfering RNA (siRNA), or over expressing ErbB4, were subjected to wound healing and apoptosis assays. RESULTS ErbB4 levels increased in Crohn's colitis and the colon epithelium of mice with DSS colitis or injected with TNF. In YAMC cells, TNF induced ErbB4 messenger RNA, protein, and phosphorylation; nuclear factor kappaB activation also stimulated ErbB4 accumulation. ErbB4 siRNA sensitized cells to TNF-stimulated apoptosis, while over expression blocked apoptosis induced by TNF plus cycloheximide. Additionally, ErbB4 siRNA decreased YAMC cell wound healing. ErbB4 knockdown attenuated, while over expression elevated, phosphorylation of Akt in response to TNF. Inhibition of the phosphatidylinositol 3-kinase/Akt signaling cascade reversed the ability of ErbB4 over expression to protect from cytokine-induced apoptosis. CONCLUSIONS ErbB4 expression and signaling are key elements for TNF responses in vivo and in cell culture, protecting intestinal epithelial cells from apoptosis in the inflammatory environment, possibly through Akt activation.
Collapse
Affiliation(s)
- Mark R. Frey
- Department of Pediatrics, Division of Gastroenterology, Hepatology & Nutrition, Vanderbilt University School of Medicine Nashville, TN 37232-0696
| | - Karen L. Edelblum
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232-0696
| | - Matthew T. Mullane
- Department of Pediatrics, Division of Gastroenterology, Hepatology & Nutrition, Vanderbilt University School of Medicine Nashville, TN 37232-0696
| | - Dongchun Liang
- Department of Pediatrics, Division of Gastroenterology, Hepatology & Nutrition, Vanderbilt University School of Medicine Nashville, TN 37232-0696
| | - D. Brent Polk
- Department of Pediatrics, Division of Gastroenterology, Hepatology & Nutrition, Vanderbilt University School of Medicine Nashville, TN 37232-0696,Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232-0696
| |
Collapse
|
49
|
Tomiya T, Omata M, Imamura H, Fujiwara K. Impaired liver regeneration in acute liver failure: the significance of cross-communication of growth associated factors in liver regeneration. Hepatol Res 2008; 38 Suppl 1:S29-33. [PMID: 19125948 DOI: 10.1111/j.1872-034x.2008.00423.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Liver regeneration is considered to be retarded or suppressed in patients with acute liver failure. However, the mechanisms for this suppression remain to be elucidated. In order to evaluate deficiencies in liver regeneration in patients with acute liver failure, we focused on the cross-communication of growth-associated factors in experimental models. In primary cultured rat hepatocytes, the levels of cellular p53 and transforming growth factor-alpha (TGF-alpha), as well as DNA synthesis increased by the addition of hepatocyte growth factor (HGF). When p53 activity was suppressed, TGF-alpha expression and DNA synthesis were reduced. DNA synthesis was also reduced when TGF-alpha activity was suppressed. In rats after partial hepatectomy, hepatic HGF and p53 protein levels increased, followed by an increase of hepatic TGF-alpha levels and hepatocyte proliferation. Circulating levels of HGF and TGF-alpha correlated with changes in the hepatic levels. The suppression of TGF-alpha activity reduced the proliferation of hepatocytes in these rats. In patients suffering from acute hepatitis, serum HGF levels increased followed by an increase of serum TGF-alpha levels. In contrast, in patients with acute liver failure, the increase of serum TGF-alpha levels was suppressed depending on the severity of hepatic failure, even though serum HGF levels markedly increased. The patients' hepatic levels were consistent with serum levels. Cross-communication of growth associated factors may be important in the progression of liver regeneration, and impaired regenerative capacity in patients with acute liver failure may be attributable, at least in part, to a disruption of communication of growth-associated factors.
Collapse
Affiliation(s)
- Tomoaki Tomiya
- Department of Gastroenterology, University of Tokyo, Tokyo, Japan
| | | | | | | |
Collapse
|
50
|
Transactivation of EGF receptor and ErbB2 protects intestinal epithelial cells from TNF-induced apoptosis. Proc Natl Acad Sci U S A 2008; 105:11772-7. [PMID: 18701712 DOI: 10.1073/pnas.0801463105] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
TNF is a pleiotropic cytokine that activates both anti- and proapoptotic signaling pathways, with cell fate determined by the balance between these two pathways. Activation of ErbB family members, including EGF receptor (EGFR/ErbB1), promotes cell survival and regulates several signals that overlap with those stimulated by TNF. This study was undertaken to determine the effects of TNF on EGFR and ErbB2 activation and intestinal epithelial cell survival. Mice, young adult mouse colon epithelial cells, and EGFR knockout mouse colon epithelial cells were treated with TNF. Activation of EGFR, ErbB2, Akt, Src, and apoptosis were determined in vivo and in vitro. TNF stimulated EGFR phosphorylation in young adult mouse colon epithelial cells, and loss of EGFR expression or inhibition of kinase activity increased TNF-induced apoptosis, which was prevented in WT but not by kinase-inactive EGFR expression. Similarly, TNF injection stimulated apoptosis in EGFR-kinase-defective mice (EGFR(wa2)) compared with WT mice. TNF also activated ErbB2, and loss of ErbB2 expression increased TNF-induced apoptosis. Furthermore, Src-kinase activity and the expression of both EGFR and ErbB2 were required for TNF-induced cell survival. Akt was shown to be a downstream target of TNF-activated EGFR and ErbB2. These findings demonstrate that EGFR and ErbB2 are critical mediators of TNF-regulated antiapoptotic signals in intestinal epithelial cells. Given evidence for TNF signaling in the development of colitis-associated carcinoma, this observation has significant implications for understanding the role of EGFR in maintaining intestinal epithelial cell homeostasis during cytokine-mediated inflammatory responses.
Collapse
|