1
|
Tawengi M, Al-Dali Y, Tawengi A, Benter IF, Akhtar S. Targeting the epidermal growth factor receptor (EGFR/ErbB) for the potential treatment of renal pathologies. Front Pharmacol 2024; 15:1394997. [PMID: 39234105 PMCID: PMC11373609 DOI: 10.3389/fphar.2024.1394997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
Epidermal growth factor receptor (EGFR), which is referred to as ErbB1/HER1, is the prototype of the EGFR family of receptor tyrosine kinases which also comprises ErbB2 (Neu, HER2), ErbB3 (HER3), and ErbB4 (HER4). EGFR, along with other ErbBs, is expressed in the kidney tubules and is physiologically involved in nephrogenesis and tissue repair, mainly following acute kidney injury. However, its sustained activation is linked to several kidney pathologies, including diabetic nephropathy, hypertensive nephropathy, glomerulonephritis, chronic kidney disease, and renal fibrosis. This review aims to provide a summary of the recent findings regarding the consequences of EGFR activation in several key renal pathologies. We also discuss the potential interplay between EGFR and the reno-protective angiotensin-(1-7) (Ang-(1-7), a heptapeptide member of the renin-angiotensin-aldosterone system that counter-regulates the actions of angiotensin II. Ang-(1-7)-mediated inhibition of EGFR transactivation might represent a potential mechanism of action for its renoprotection. Our review suggests that there is a significant body of evidence supporting the potential inhibition of EGFR/ErbB, and/or administration of Ang-(1-7), as potential novel therapeutic strategies in the treatment of renal pathologies. Thus, EGFR inhibitors such as Gefitinib and Erlinotib that have an acceptable safety profile and have been clinically used in cancer chemotherapy since their FDA approval in the early 2000s, might be considered for repurposing in the treatment of renal pathologies.
Collapse
Affiliation(s)
- Mohamed Tawengi
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Yazan Al-Dali
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Ibrahim F Benter
- Faculty of Pharmacy, Final International University, Kyrenia, Cyprus
| | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
2
|
Gekle M, Dubourg V, Schwerdt G, Benndorf RA, Schreier B. The role of EGFR in vascular AT1R signaling: From cellular mechanisms to systemic relevance. Biochem Pharmacol 2023; 217:115837. [PMID: 37777161 DOI: 10.1016/j.bcp.2023.115837] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
The epidermal growth factor receptor (EGFR) belongs to the ErbB-family of receptor tyrosine kinases that are of importance in oncology. During the last years, substantial evidence accumulated for a crucial role of EGFR concerning the action of the angiotensin II type 1 receptor (AT1R) in blood vessels, resulting form AT1R-induced EGFR transactivation. This transactivation occurs through the release of membrane-anchored EGFR-ligands, cytosolic tyrosine kinases, heterocomplex formation or enhanced ligand expression. AT1R-EGFR crosstalk amplifies the signaling response and enhances the biological effects of angiotensin II. Downstream signaling cascades include ERK1/2 and p38 MAPK, PLCγ and STAT. AT1R-induced EGFR activation contributes to vascular remodeling and hypertrophy via e.g. smooth muscle cell proliferation, migration and extracellular matrix production. EGFR transactivation results in increased vessel wall thickness and reduced vascular compliance. AT1R and EGFR signaling pathways are also implicated the induction of vascular inflammation. Again, EGFR transactivation exacerbates the effects, leading to endothelial dysfunction that contributes to vascular inflammation, dysfunction and remodeling. Dysregulation of the AT1R-EGFR axis has been implicated in the pathogenesis of various cardiovascular diseases and inhibition or prevention of EGFR signaling can attenuate part of the detrimental impact of enhanced renin-angiotensin-system (RAAS) activity, highlighting the importance of EGFR for the adverse consequences of AT1R activation. In summary, EGFR plays a critical role in vascular AT1R action, enhancing signaling, promoting remodeling, contributing to inflammation, and participating in the pathogenesis of cardiovascular diseases. Understanding the interplay between AT1R and EGFR will foster the development of effective therapeutic strategies of RAAS-induced disorders.
Collapse
Affiliation(s)
- Michael Gekle
- Julius-Bernstein-Institute of Physiology, Martin-Luther-University Halle-Wittenberg, Magdeburger Str. 6, D-06112 Halle (Saale), Germany.
| | - Virginie Dubourg
- Julius-Bernstein-Institute of Physiology, Martin-Luther-University Halle-Wittenberg, Magdeburger Str. 6, D-06112 Halle (Saale), Germany
| | - Gerald Schwerdt
- Julius-Bernstein-Institute of Physiology, Martin-Luther-University Halle-Wittenberg, Magdeburger Str. 6, D-06112 Halle (Saale), Germany
| | - Ralf A Benndorf
- Institute of Pharmacy, Martin-Luther-University, Halle, Germany
| | - Barbara Schreier
- Julius-Bernstein-Institute of Physiology, Martin-Luther-University Halle-Wittenberg, Magdeburger Str. 6, D-06112 Halle (Saale), Germany
| |
Collapse
|
3
|
Schreier B, Stern C, Rabe S, Mildenberger S, Gekle M. Assessment of the Role of Endothelial and Vascular Smooth Muscle EGFR for Acute Blood Pressure Effects of Angiotensin II and Adrenergic Stimulation in Obese Mice. Biomedicines 2023; 11:2241. [PMID: 37626737 PMCID: PMC10452314 DOI: 10.3390/biomedicines11082241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/05/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
(1) Background: Obesity is associated with hypertension because of endocrine dysregulation of the adrenergic and the renin-angiotensin-aldosterone systems. The epidermal growth factor receptor (EGFR) is an important signaling hub in the cardiovascular system. In this study, we investigate the role of smooth muscle cell (VSMC) and endothelial cell (EC) EGFRs for blood pressure homeostasis and acute vascular reactivity in vivo. (2) Methods: Mice with deletion of the EGFR in the respective cell type received either a high-fat (HFD) or standard-fat diet (SFD) for 18 weeks. Intravascular blood pressure was measured via a Millar catheter in anesthetized animals upon vehicle load, angiotensin II (AII) and phenylephrine (PE) stimulation. (3) Results: We confirmed that deletion of the EGFR in VSMCs leads to reduced blood pressure and a most probably compensatory heart rate increase. EC-EGFR and VSMC-EGFR had only a minor impact on volume-load-induced blood pressure changes in lean as well as in obese wild-type animals. Regarding vasoactive substances, EC-EGFR seems to have no importance for angiotensin II action and counteracting HFD-induced prolonged blood pressure increase upon PE stimulation. VSMC-EGFR supports the blood pressure response to adrenergic and angiotensin II stimulation in lean animals. The responsiveness to AII and alpha-adrenergic stimulation was similar in lean and obese animals despite the known enhanced activity of the RAAS and the sympathetic nervous system under a high-fat diet. (4) Conclusions: We demonstrate that EGFRs in VSMCs and to a lesser extent in ECs modulate short-term vascular reactivity to AII, catecholamines and volume load in lean and obese animals.
Collapse
Affiliation(s)
- Barbara Schreier
- Julius-Bernstein-Institute of Physiology, Martin-Luther-University Halle-Wittenberg, 06112 Halle, Germany
| | | | | | | | | |
Collapse
|
4
|
Sun F, Liu J, Wang Y, Yang H, Song D, Fu H, Feng X. BASP1 promotes high glucose-induced endothelial apoptosis in diabetes via activation of EGFR signaling. J Diabetes Investig 2023; 14:535-547. [PMID: 36756695 PMCID: PMC10034959 DOI: 10.1111/jdi.13920] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/08/2022] [Accepted: 09/19/2022] [Indexed: 02/10/2023] Open
Abstract
AIMS Diabetes mellitus is a common chronic disease of glucose metabolism. Endothelial dysfunction is an early event in diabetes complicated by cardiovascular disease. This study aimed to reveal the expression of BASP1 and its biological roles in endothelial cell dysfunction in diabetes complicated by cardiovascular disease. MATERIALS AND METHODS By analyzing the databases related to diabetes complicated with coronary heart disease, BASP1 was screened out as an upregulated gene. Human umbilical vein endothelial cells (HUVECs) and primary mouse aortic endothelial cells were treated with high glucose to establish cell models of diabetes-related endothelial dysfunction, and the expression changes of BASP1 were verified by RT-qPCR, western blot, and immunofluorescence. BASP1 was silenced or overexpressed by siRNA or overexpression plasmid, and its effects on cell migration, apoptosis, tube formation, inflammatory response, and ROS were detected. The possible signaling pathway of BASP1 was found and the mechanism of BASP1 on promoting the progression of endothelial dysfunction was explored using the EGFR inhibitor, gefitinib. RESULTS Bioinformatics analysis indicated that the expression of BASP1 in patients with diabetes mellitus and concomitant coronary heart disease was increased. High glucose induced the upregulation of BASP1 expression in endothelial cells, and showed a time-dependent relationship. Silencing of BASP1 alleviated the damage of high glucose to endothelial cells. BASP1 regulated EGFR positively. The promoting effect of BASP1 on endothelial cell apoptosis may be achieved by regulating the EGFR pathway. CONCLUSION BASP1 promotes endothelial cell injury induced by high glucose in patients with diabetes, which may be activated by activating the EGFR pathway.
Collapse
Affiliation(s)
- Fengnan Sun
- Department of Laboratory Medicine, Yantaishan Hospital, Yantai, China
| | - Junwei Liu
- Department of Laboratory Medicine, Qishan Hospital, Yantai, China
| | - Yanzheng Wang
- Department of Laboratory Medicine, Yantaishan Hospital, Yantai, China
| | - Hongmei Yang
- Department of Laboratory Medicine, Yantaishan Hospital, Yantai, China
| | - Danfeng Song
- Department of Laboratory Medicine, Yantaishan Hospital, Yantai, China
| | - Haiyan Fu
- Department of Laboratory Medicine, Yantaishan Hospital, Yantai, China
| | - Xingxing Feng
- Kunming Key Laboratory of Children Infection and Immunity, Yunnan Key Laboratory of Children's Major Disease Research, Yunnan Institute of Pediatrics, Kunming Children's Hospital, Kunming, China
| |
Collapse
|
5
|
Shoykhet M, Dervishi O, Menauer P, Hiermaier M, Moztarzadeh S, Osterloh C, Ludwig RJ, Williams T, Gerull B, Kääb S, Clauss S, Schüttler D, Waschke J, Yeruva S. EGFR inhibition leads to enhanced desmosome assembly and cardiomyocyte cohesion via ROCK activation. JCI Insight 2023; 8:163763. [PMID: 36795511 PMCID: PMC10070108 DOI: 10.1172/jci.insight.163763] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/15/2023] [Indexed: 02/17/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a familial heart disease partly caused by impaired desmosome turnover. Thus, stabilization of desmosome integrity may provide new treatment options. Desmosomes, apart from cellular cohesion, provide the structural framework of a signaling hub. Here, we investigated the role of the epidermal growth factor receptor (EGFR) in cardiomyocyte cohesion. We inhibited EGFR under physiological and pathophysiological conditions using the murine plakoglobin-KO AC model, in which EGFR was upregulated. EGFR inhibition enhanced cardiomyocyte cohesion. Immunoprecipitation showed an interaction of EGFR and desmoglein 2 (DSG2). Immunostaining and atomic force microscopy (AFM) revealed enhanced DSG2 localization and binding at cell borders upon EGFR inhibition. Enhanced area composita length and desmosome assembly were observed upon EGFR inhibition, confirmed by enhanced DSG2 and desmoplakin (DP) recruitment to cell borders. PamGene Kinase assay performed in HL-1 cardiomyocytes treated with erlotinib, an EGFR inhibitor, revealed upregulation of Rho-associated protein kinase (ROCK). Erlotinib-mediated desmosome assembly and cardiomyocyte cohesion were abolished upon ROCK inhibition. Thus, inhibiting EGFR and, thereby, stabilizing desmosome integrity via ROCK might provide treatment options for AC.
Collapse
Affiliation(s)
- Maria Shoykhet
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Orsela Dervishi
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Philipp Menauer
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Matthias Hiermaier
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Sina Moztarzadeh
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Colin Osterloh
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Tatjana Williams
- Comprehensive Heart Failure Center and Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Brenda Gerull
- Comprehensive Heart Failure Center and Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Stefan Kääb
- Medizinische Klinik und Poliklinik I, LMU Hospital, LMU, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modeling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Sebastian Clauss
- Medizinische Klinik und Poliklinik I, LMU Hospital, LMU, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modeling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, LMU Hospital, LMU, Munich, Germany
| | - Dominik Schüttler
- Medizinische Klinik und Poliklinik I, LMU Hospital, LMU, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modeling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, LMU Hospital, LMU, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Sunil Yeruva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| |
Collapse
|
6
|
Morita D, Ito K, Ikeuchi N, Nishida Y, Igata F, Nakamura T, Murayama H, Watanabe M, Takahashi K, Yasuno T, Uesugi N, Fujita M, Oda T, Masutani K. A case of crescentic glomerulonephritis induced by afatinib for lung adenocarcinoma. CEN Case Rep 2022; 12:152-158. [PMID: 36180718 PMCID: PMC10151294 DOI: 10.1007/s13730-022-00737-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 09/15/2022] [Indexed: 10/14/2022] Open
Abstract
Afatinib is a second-generation, oral, epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI). One of the most common adverse effects of affatinib is diarrhea, which may lead to acute kidney injury (AKI) due to severe plasma volume loss; however, no case of glomerular injury directly induced by afatinib has been reported to date. Here, we describe the case of a 53-year-old Japanese male patient with advanced lung adenocarcinoma who twice developed AKI requiring dialysis, once after starting and once after increasing the dose of afatinib. Although serum anti-neutrophil cytoplasmic antibodies were negative, crescentic glomerulonephritis with no immune deposits was confirmed on kidney biopsy. No vasculitis-like signs were observed in other organs, such as lung, skin, or peripheral nerves. Afatinib was considered the cause of glomerular damage and was immediately discontinued; corticosteroids were administered. Renal function gradually recovered thereafter, with serum creatinine levels at ~ 2.3 mg/dL after second-line therapy with bevacizumab and atezolizumab. Several cases of cutaneous leukocytoclastic vasculitis have been reported in patients treated with other EGFR-TKIs; therefore, afatinib-induced vasculitis may lead to crescentic glomerulonephritis. Although afatinib-induced glomerular injury is extremely rare and has an unclear mechanism, renal function and urinary findings need to be closely monitored.
Collapse
|
7
|
Méndez-Morales S, Pérez-De Marco J, Rodríguez-Cortés O, Flores-Mejía R, Martínez-Venegas M, Sánchez-Vera Y, Tamay-Cach F, Lomeli-Gonzaléz J, Emilio Reyes A, Lehman-Mendoza R, Martínez-Arredondo H, Vazquez-Dávila R, Torres-Roldan J, Correa-Basurto J, Arellano-Mendoza M. Diabetic neuropathy: Molecular approach a treatment opportunity. Vascul Pharmacol 2022; 143:106954. [PMID: 35063655 DOI: 10.1016/j.vph.2022.106954] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 12/15/2022]
|
8
|
Shraim BA, Moursi MO, Benter IF, Habib AM, Akhtar S. The Role of Epidermal Growth Factor Receptor Family of Receptor Tyrosine Kinases in Mediating Diabetes-Induced Cardiovascular Complications. Front Pharmacol 2021; 12:701390. [PMID: 34408653 PMCID: PMC8365470 DOI: 10.3389/fphar.2021.701390] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022] Open
Abstract
Diabetes mellitus is a major debilitating disease whose global incidence is progressively increasing with currently over 463 million adult sufferers and this figure will likely reach over 700 million by the year 2045. It is the complications of diabetes such as cardiovascular, renal, neuronal and ocular dysfunction that lead to increased patient morbidity and mortality. Of these, cardiovascular complications that can result in stroke and cardiomyopathies are 2- to 5-fold more likely in diabetes but the underlying mechanisms involved in their development are not fully understood. Emerging research suggests that members of the Epidermal Growth Factor Receptor (EGFR/ErbB/HER) family of tyrosine kinases can have a dual role in that they are beneficially required for normal development and physiological functioning of the cardiovascular system (CVS) as well as in salvage pathways following acute cardiac ischemia/reperfusion injury but their chronic dysregulation may also be intricately involved in mediating diabetes-induced cardiovascular pathologies. Here we review the evidence for EGFR/ErbB/HER receptors in mediating these dual roles in the CVS and also discuss their potential interplay with the Renin-Angiotensin-Aldosterone System heptapeptide, Angiotensin-(1-7), as well the arachidonic acid metabolite, 20-HETE (20-hydroxy-5, 8, 11, 14-eicosatetraenoic acid). A greater understanding of the multi-faceted roles of EGFR/ErbB/HER family of tyrosine kinases and their interplay with other key modulators of cardiovascular function could facilitate the development of novel therapeutic strategies for treating diabetes-induced cardiovascular complications.
Collapse
Affiliation(s)
- Bara A Shraim
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Moaz O Moursi
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Ibrahim F Benter
- Faculty of Medicine, Eastern Mediterranean University, Famagusta, North Cyprus
| | - Abdella M Habib
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
9
|
Endothelial epidermal growth factor receptor is of minor importance for vascular and renal function and obesity-induced dysfunction in mice. Sci Rep 2021; 11:7269. [PMID: 33790318 PMCID: PMC8012653 DOI: 10.1038/s41598-021-86587-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 03/16/2021] [Indexed: 12/11/2022] Open
Abstract
Vascular EGF receptors (EGFR) influence function and structure of arterial vessels. In genetic mouse models we described the role of vascular smooth muscle (VSMC) EGFR for proper physiological function and structure as well as for pathophysiological alterations by obesity or angiotensin II. As the importance of endothelial (EC) EGFR in vivo is unknown, we analyzed the impact of EC-EGFR knockout in a conditional mouse model on vascular and renal function under control condition as well as in obesity and in comparison to VSMC-KO. Heart and lung weight, blood pressure and aortic transcriptome (determined by RNA-seq) were not affected by EC-EGFR-KO. Aortic reactivity to α1-adrenergic stimulation was not affected by EC-EGFR-KO contrary to VSMC-EGFR-KO. Endothelial-induced relaxation was reduced in abdominal aorta of EC-EGFR-KO animals, whereas it was enhanced in VSMC-EGFR-KO animals. Mesenteric arteries of EC-EGFR-KO animals showed enhanced sensitivity to α1-adrenergic stimulation, whereas endothelial-induced relaxation and vessel morphology were not affected. Renal weight, histomorphology, function (albumin excretion, serum creatinine, fractional water excretion) or transcriptome were not affected by EC-EGFR-KO, likewise in VSMC-EGFR-KO. High fat diet (HFD) over 18 weeks induced arterial wall thickening, renal weight increase, creatininemia, renal and aortic transcriptome alterations with a similar pattern in EC-EGFR-WT and EC-EGFR-KO animals by contrast to the previously reported impact of VSMC-EGFR-KO. HFD induced endothelial dysfunction in abdominal aortae of EC-EGFR-WT, which was not additive to the EC-EGFR-KO-induced endothelial dysfunction. As shown before, VSMC-EGFR-KO prevented HFD-induced endothelial dysfunction. HFD-induced albuminuria was less pronounced in EC-EGFR-KO animals and abrogated in VSMC-EGFR-KO animals. Our results indicate that EC-EGFR, in comparison to VSMC-EGFR, is of minor and opposite importance for basal renovascular function as well as for high fat diet-induced vascular remodeling and renal end organ damage.
Collapse
|
10
|
Sheng L, Bayliss G, Zhuang S. Epidermal Growth Factor Receptor: A Potential Therapeutic Target for Diabetic Kidney Disease. Front Pharmacol 2021; 11:598910. [PMID: 33574751 PMCID: PMC7870700 DOI: 10.3389/fphar.2020.598910] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetic kidney disease (DKD) is a leading cause of end-stage renal disease worldwide and the major cause of renal failure among patients on hemodialysis. Numerous studies have demonstrated that transient activation of epidermal growth factor receptor (EGFR) pathway is required for promoting kidney recovery from acute injury whereas its persistent activation is involved in the progression of various chronic kidney diseases including DKD. EGFR-mediated pathogenesis of DKD is involved in hemodynamic alteration, metabolic disturbance, inflammatory response and parenchymal cellular dysfunction. Therapeutic intervention of this receptor has been available in the oncology setting. Targeting EGFR might also hold a therapeutic potential for DKD. Here we review the functional role of EGFR in the development of DKD, mechanisms involved and the perspective about use of EGFR inhibitors as a treatment for DKD.
Collapse
Affiliation(s)
- Lili Sheng
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - George Bayliss
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, United States
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, United States
| |
Collapse
|
11
|
Stern C, Schreier B, Nolze A, Rabe S, Mildenberger S, Gekle M. Knockout of vascular smooth muscle EGF receptor in a mouse model prevents obesity-induced vascular dysfunction and renal damage in vivo. Diabetologia 2020; 63:2218-2234. [PMID: 32548701 PMCID: PMC7476975 DOI: 10.1007/s00125-020-05187-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 04/06/2020] [Indexed: 12/17/2022]
Abstract
AIMS/HYPOTHESIS Obesity causes type 2 diabetes leading to vascular dysfunction and finally renal end-organ damage. Vascular smooth muscle (VSM) EGF receptor (EGFR) modulates vascular wall homeostasis in part via serum response factor (SRF), a major regulator of VSM differentiation and a sensor for glucose. We investigated the role of VSM-EGFR during obesity-induced renovascular dysfunction, as well as EGFR-hyperglycaemia crosstalk. METHODS The role of VSM-EGFR during high-fat diet (HFD)-induced type 2 diabetes was investigated in a mouse model with inducible, VSM-specific EGFR-knockout (KO). Various structural and functional variables as well as transcriptome changes, in vivo and ex vivo, were assessed. The impact of hyperglycaemia on EGFR-induced signalling and SRF transcriptional activity and the underlying mechanisms were investigated at the cellular level. RESULTS We show that VSM-EGFR mediates obesity/type 2 diabetes-induced vascular dysfunction, remodelling and transcriptome dysregulation preceding renal damage and identify an EGFR-glucose synergism in terms of SRF activation, matrix dysregulation and mitochondrial function. EGFR deletion protects the animals from HFD-induced endothelial dysfunction, creatininaemia and albuminuria. Furthermore, we show that HFD leads to marked changes of the aortic transcriptome in wild-type but not in KO animals, indicative of EGFR-dependent SRF activation, matrix dysregulation and mitochondrial dysfunction, the latter confirmed at the cellular level. Studies at the cellular level revealed that high glucose potentiated EGFR/EGF receptor 2 (ErbB2)-induced stimulation of SRF activity, enhancing the graded signalling responses to EGF, via the EGFR/ErbB2-ROCK-actin-MRTF pathway and promoted mitochondrial dysfunction. CONCLUSIONS/INTERPRETATION VSM-EGFR contributes to HFD-induced vascular and subsequent renal alterations. We propose that a potentiated EGFR/ErbB2-ROCK-MRTF-SRF signalling axis and mitochondrial dysfunction underlie the role of EGFR. This advanced working hypothesis will be investigated in mechanistic depth in future studies. VSM-EGFR may be a therapeutic target in cases of type 2 diabetes-induced renovascular disease. DATA AVAILABILITY The datasets generated during and/or analysed during the current study are available in: (1) share_it, the data repository of the academic libraries of Saxony-Anhalt ( https://doi.org/10.25673/32049.2 ); and (2) in the gene expression omnibus database with the study identity GSE144838 ( https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE144838 ). Graphical abstract.
Collapse
Affiliation(s)
- Christian Stern
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany
| | - Barbara Schreier
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany
| | - Alexander Nolze
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany
| | - Sindy Rabe
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany
| | - Sigrid Mildenberger
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany
| | - Michael Gekle
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany.
| |
Collapse
|
12
|
Habib AM, Nagi K, Thillaiappan NB, Sukumaran V, Akhtar S. Vitamin D and Its Potential Interplay With Pain Signaling Pathways. Front Immunol 2020; 11:820. [PMID: 32547536 PMCID: PMC7270292 DOI: 10.3389/fimmu.2020.00820] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 04/09/2020] [Indexed: 12/12/2022] Open
Abstract
About 50 million of the U.S. adult population suffer from chronic pain. It is a complex disease in its own right for which currently available analgesics have been deemed woefully inadequate since ~20% of the sufferers derive no benefit. Vitamin D, known for its role in calcium homeostasis and bone metabolism, is thought to be of clinical benefit in treating chronic pain without the side-effects of currently available analgesics. A strong correlation between hypovitaminosis D and incidence of bone pain is known. However, the potential underlying mechanisms by which vitamin D might exert its analgesic effects are poorly understood. In this review, we discuss pathways involved in pain sensing and processing primarily at the level of dorsal root ganglion (DRG) neurons and the potential interplay between vitamin D, its receptor (VDR) and known specific pain signaling pathways including nerve growth factor (NGF), glial-derived neurotrophic factor (GDNF), epidermal growth factor receptor (EGFR), and opioid receptors. We also discuss how vitamin D/VDR might influence immune cells and pain sensitization as well as review the increasingly important topic of vitamin D toxicity. Further in vitro and in vivo experimental studies will be required to study these potential interactions specifically in pain models. Such studies could highlight the potential usefulness of vitamin D either alone or in combination with existing analgesics to better treat chronic pain.
Collapse
Affiliation(s)
| | | | | | | | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
13
|
Wang ZJ, Chang LL, Wu J, Pan HM, Zhang QY, Wang MJ, Xin XM, Luo SS, Chen JA, Gu XF, Guo W, Zhu YZ. A Novel Rhynchophylline Analog, Y396, Inhibits Endothelial Dysfunction Induced by Oxidative Stress in Diabetes Through Epidermal Growth Factor Receptor. Antioxid Redox Signal 2020; 32:743-765. [PMID: 31892280 DOI: 10.1089/ars.2018.7721] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Aims: Endothelial dysfunction appears in early diabetes mellitus partially because of epidermal growth factor receptor (EGFR) abnormal activation and downstream oxidative stress. The aim of this study was to determine whether Y396, a synthesized analog of rhynchophylline, could protect against endothelial dysfunction in diabetes and the underlying molecular mechanism. Results: Y396 could directly target the EGFR and inhibit its phosphorylation induced by high glucose and EGF, downstream translocation to the nucleus of E2F1, and its transcriptional activity and expression of Nox4. Diabetes-induced endothelium malfunction was ameliorated by Y396 treatment through EGFR inhibition. Downstream oxidative stress was decreased by Y396 in the aortas of type 1 diabetes mellitus mice and primary rat aorta endothelial cells (RAECs). Y396 could also ameliorate tunicamycin-induced oxidative stress in the aorta and RAECs. In addition, we again determined the protective effects of Y396 on high-fat diet/streptozotocin-induced type 2 diabetes mellitus. Innovation: This is the first study to demonstrate that Y396, a novel rhynchophylline analog, suppressed high-glucose-induced endothelial malfunction both in vivo and in vitro by inhibiting abnormal phosphorylation of EGFR. Our work uncovered EGFR as a novel therapeutic target and Y396 as a potential therapy against diabetes-induced complication. Conclusion: Y396 could directly bind with EGFR, and inhibit its phosphorylation and downstream E2F1 transcriptional activity. It could also preserve tunicamycin-evoked endothelial dysfunction and oxidative stress. It could protect against diabetes-induced endothelium malfunction in vivo through EGFR inhibition and downstream oxidative stress. Antioxid. Redox Signal. 32, 743-765.
Collapse
Affiliation(s)
- Zhi-Jun Wang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China.,State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| | - Ling-Ling Chang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Jian Wu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Hong-Ming Pan
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Qiu-Yan Zhang
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, China
| | - Min-Jun Wang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Xiao-Ming Xin
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Shan-Shan Luo
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Ji-An Chen
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Xian-Feng Gu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Wei Guo
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Yi-Zhun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China.,State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| |
Collapse
|
14
|
Li J, Huang Y, Zhao S, Guo Q, Zhou J, Han W, Xu Y. Based on network pharmacology to explore the molecular mechanisms of astragalus membranaceus for treating T2 diabetes mellitus. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:633. [PMID: 31930034 DOI: 10.21037/atm.2019.10.118] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Astragalus membranaceus refers to a type of traditional Chinese medicine (TCM) used to treat type 2 diabetes mellitus (T2DM), whereas its molecular mechanism remains unclear. In the presented study, network pharmacology was performed to analyze the molecular mechanism of astragalus membranaceus against T2DM. Methods First, we found common targets of astragalus membranaceus and disease, protein-protein interaction (PPI) network was built by String, and then key targets were screened from these common targets by topological analysis. Subsequently, common targets were introduced into DAVID to achieve the results of gene ontology (GO) and KEGG enrichment analysis. The therapeutic effect of astragalus was observed, and several key targets were verified by an animal experiment. Results First, 13 key targets (EGFR, KDR, SRC, ERBB2, FYN, ESR1, AR, HSP90AA1, PTGS2, ABCG2, AB1, MMP2, and CYP1) were found by topological analysis. Then, the results of GO and KEGG suggested that the anti-diabetes effect of astragalus membranaceus was strongly associated with the activation of receptor protein tyrosine kinase (RPTK). The results of animal experiments revealed that astragalus could enhance the morphology of rat pancreas and up-regulate the expression of tyrosine receptor. Conclusions In brief, 13 key targets were found in this study, and astragalus membranaceus was found up-regulating insulin signaling pathways by improving the activity of casein kinase, regulating lipid metabolism, and enhancing insulin resistance to treat T2DM. The present study lays a basis for subsequent experimental research and broadens the clinical application of astragalus membranaceus.
Collapse
Affiliation(s)
- Jie Li
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yanqin Huang
- Department of Endocrine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Sen Zhao
- Department of Chinese Medicine, The General Hospital of the People's Liberation Army, Beijing 100853, China
| | - Qiuyue Guo
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jie Zhou
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Wenjing Han
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yunsheng Xu
- Department of Endocrine, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250001, China
| |
Collapse
|
15
|
Zhang W, Yang H, Zhu L, Luo Y, Nie L, Li G. Role of EGFR/ErbB2 and PI 3K/AKT/e-NOS in Lycium barbarum polysaccharides Ameliorating Endothelial Dysfunction Induced by Oxidative Stress. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:1523-1539. [PMID: 31645123 DOI: 10.1142/s0192415x19500782] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lycium barbarum polysaccharides (LBP) are the major ingredients of wolfberry. In this study, we investigated the role of LBP in endothelial dysfunction induced by oxidative stress and the underlying mechanisms using thoracic aortic endothelial cells of rat (RAECs) as a model. We found that Ang II inhibits cell viability of RAECs with 10-6mol/L of Ang II treatment for 24h most potential (P<0.05), the level of reactive oxygen species (ROS) is increased by Ang II treatment (P<0.01), and the expression of Occludin and Zonula occludens-1 (ZO-1) is decreased by Ang II treatment (P<0.05). However, preincubation of cells with LBP could inhibit the changes caused by Ang II, LBP increased cell viability (P<0.05), decreased the level of ROS (P<0.01), and up-regulated the expression of Occludin (P<0.05) and ZO-1. In addition, Ang II treatment increased the expression of EGFR and p-EGFR (Try1172) and which can be inhibited by LBP. On the contrary, expression of ErbB2, p-ErbB2 (Try1248), PI3K, p-e-NOS (Ser1177) (P<0.05), and p-AKT (Ser473) (P<0.05) was inhibited by Ang II treatment and which can be increased by LBP. Treatment of the cells with inhibitors showed that the regulation of p-e-NOS and p-AKT expression by Ang II and LBP can be blocked by PI3K inhibitor wortmannin but not EGFR and ErbB2 inhibitor AC480. Taken together, our results suggested that LBP plays a critical role in maintaining the integrality of blood vessel endothelium through reduced production of ROS via regulating the activity of EGFR, ErbB2, PI3K/AKT/e-NOS, and which may offer a novel therapeutic option in the management of endothelial dysfunction.
Collapse
Affiliation(s)
- Wenjuan Zhang
- School of Public Health and Management, Ningxia Medical University, Shengli Street 1160, Yinchuan 750004, P. R. China
| | - Huifang Yang
- School of Public Health and Management, Ningxia Medical University, Shengli Street 1160, Yinchuan 750004, P. R. China
| | - Lingqin Zhu
- School of Public Health and Management, Ningxia Medical University, Shengli Street 1160, Yinchuan 750004, P. R. China
| | - Yan Luo
- School of Basic Medical Science, Ningxia Medical University, Shengli Street 1160, Yinchuan 750004, P. R. China
| | - Lihong Nie
- School of Basic Medical Science, Ningxia Medical University, Shengli Street 1160, Yinchuan 750004, P. R. China
| | - Guanghua Li
- School of Basic Medical Science, Ningxia Medical University, Shengli Street 1160, Yinchuan 750004, P. R. China.,School of Public Health and Management, Ningxia Medical University, Shengli Street 1160, Yinchuan 750004, P. R. China
| |
Collapse
|
16
|
Role of Epidermal Growth Factor Receptor (EGFR) and Its Ligands in Kidney Inflammation and Damage. Mediators Inflamm 2018; 2018:8739473. [PMID: 30670929 PMCID: PMC6323488 DOI: 10.1155/2018/8739473] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/29/2018] [Accepted: 11/07/2018] [Indexed: 12/29/2022] Open
Abstract
Chronic kidney disease (CKD) is characterized by persistent inflammation and progressive fibrosis, ultimately leading to end-stage renal disease. Although many studies have investigated the factors involved in the progressive deterioration of renal function, current therapeutic strategies only delay disease progression, leaving an unmet need for effective therapeutic interventions that target the cause behind the inflammatory process and could slow down or reverse the development and progression of CKD. Epidermal growth factor receptor (EGFR) (ERBB1), a membrane tyrosine kinase receptor expressed in the kidney, is activated after renal damage, and preclinical studies have evidenced its potential as a therapeutic target in CKD therapy. To date, seven official EGFR ligands have been described, including epidermal growth factor (EGF) (canonical ligand), transforming growth factor-α, heparin-binding epidermal growth factor, amphiregulin, betacellulin, epiregulin, and epigen. Recently, the connective tissue growth factor (CTGF/CCN2) has been described as a novel EGFR ligand. The direct activation of EGFR by its ligands can exert different cellular responses, depending on the specific ligand, tissue, and pathological condition. Among all EGFR ligands, CTGF/CCN2 is of special relevance in CKD. This growth factor, by binding to EGFR and downstream signaling pathway activation, regulates renal inflammation, cell growth, and fibrosis. EGFR can also be “transactivated” by extracellular stimuli, including several key factors involved in renal disease, such as angiotensin II, transforming growth factor beta (TGFB), and other cytokines, including members of the tumor necrosis factor superfamily, showing another important mechanism involved in renal pathology. The aim of this review is to summarize the contribution of EGFR pathway activation in experimental kidney damage, with special attention to the regulation of the inflammatory response and the role of some EGFR ligands in this process. Better insights in EGFR signaling in renal disease could improve our current knowledge of renal pathology contributing to therapeutic strategies for CKD development and progression.
Collapse
|
17
|
Identifying pathways modulating sleep duration: from genomics to transcriptomics. Sci Rep 2017; 7:4555. [PMID: 28676676 PMCID: PMC5496883 DOI: 10.1038/s41598-017-04027-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/10/2017] [Indexed: 01/02/2023] Open
Abstract
Recognizing that insights into the modulation of sleep duration can emerge by exploring the functional relationships among genes, we used this strategy to explore the genome-wide association results for this trait. We detected two major signalling pathways (ion channels and the ERBB signalling family of tyrosine kinases) that could be replicated across independent GWA studies meta-analyses. To investigate the significance of these pathways for sleep modulation, we performed transcriptome analyses of short sleeping flies’ heads (knockdown for the ABCC9 gene homolog; dSur). We found significant alterations in gene-expression in the short sleeping knockdowns versus controls flies, which correspond to pathways associated with sleep duration in our human studies. Most notably, the expression of Rho and EGFR (members of the ERBB signalling pathway) genes was down- and up-regulated, respectively, consistently with the established role of these genes for sleep consolidation in Drosophila. Using a disease multifactorial interaction network, we showed that many of the genes of the pathways indicated to be relevant for sleep duration had functional evidence of their involvement with sleep regulation, circadian rhythms, insulin secretion, gluconeogenesis and lipogenesis.
Collapse
|
18
|
Transactivation of the epidermal growth factor receptor in responses to myocardial stress and cardioprotection. Int J Biochem Cell Biol 2017; 83:97-110. [PMID: 28049018 DOI: 10.1016/j.biocel.2016.12.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/25/2016] [Accepted: 12/26/2016] [Indexed: 12/20/2022]
|
19
|
Tang X, Qin Q, Xie X, He P. Protective effect of sRAGE on fetal development in pregnant rats with gestational diabetes mellitus. Cell Biochem Biophys 2016; 71:549-56. [PMID: 25205260 DOI: 10.1007/s12013-014-0233-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
To investigate the protective effect of secretory receptor for advanced glycation endproducts (sRAGE) on the fetal development using rat model of gestational diabetes mellitus (GDM). The model of pregnant rats with intrauterine hyperglycemia was established by intraperitoneal injection of 25 mg/kg streptozotocin (STZ). Rats with established GDM were randomly grouped, and the pregnant rats in the experimental group were subsequently injected with recombinant sRAGE protein (5 mg/kg, in 0.2 mL PBS) at tail vein every 24 h, while the rats in control group were injected with the same dosage of albumin solution. Blood glucose, serum levels of advanced glycation endproducts (AGEs), and levels of RAGE protein in brain and heart tissues of pregnant rats were measured at 3, 13, and 19 days postconception. At 19 days fetuses were delivered by cesarean section, number of fetuses, their weight and placental weights were recorded, and fetal malformations and defects were analyzed visually and pathologically. The expression level of RAGE, NOX2, MCP-1, p65, VCAM-1, and VEGF mRNA in placenta was evaluated by real-time PCR. p65 protein localization was detected by immunohistochemistry in fetal brain and heart tissue sections. We analyzed the correlation between AGEs and RAGE level and the development of fetal rats, and the protective effect of blocking AGEs-RAGE pathway on the fetal development in the rat model of GDM was investigated. (1) The concentration of blood glucose and AGEs in serum of pregnant rats with GDM was significantly higher than in control group (p < 0.05), with strong correlation between blood glucose and levels of AGEs (r = 0.693, p < 0.05). (2) While both the number of fetuses and placental wet weight in pregnant rat model of GDM were similar to control group, pups from GDM group exhibited higher incidence of developmental abnormalities and higher average weight (p < 0.05). sRAGE treatment slightly but not significantly reduced the probability of the fetal developmental defects, as compared to GDM group. (3) p65, a part of the NF-kB heterodimeric complex, was localized to cell nuclei in the fetal tissues of pups delivered by GDM rats, while sRAGE treatment partially restored cytoplasmic localization of p65, similarly to control tissues. Increased incidence of fetal developmental defects observed in offsprings of pregnant rats with GDM had significant correlation with the level of AGEs in serum of pregnant rats and expression levels of RAGE protein in tissues. GDM resulted in upregulation of mRNA expression of several pro-inflammatory and ROS-inducing genes in placental tissues of pregnant rats. Elevated blood glucose, serum AGEs levels, and increased gene expression are attenuated by intravenous sRAGE treatment. sRAGE appears to reduce the activity of NF-κB in fetal tissues, thus potentially having a protective effect on fetal development.
Collapse
Affiliation(s)
- Xuwen Tang
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, China,
| | | | | | | |
Collapse
|
20
|
Akhtar S, Chandrasekhar B, Attur S, Dhaunsi GS, Yousif MHM, Benter IF. Transactivation of ErbB Family of Receptor Tyrosine Kinases Is Inhibited by Angiotensin-(1-7) via Its Mas Receptor. PLoS One 2015; 10:e0141657. [PMID: 26536590 PMCID: PMC4633289 DOI: 10.1371/journal.pone.0141657] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/12/2015] [Indexed: 01/03/2023] Open
Abstract
Transactivation of the epidermal growth factor receptor (EGFR or ErbB) family members, namely EGFR and ErbB2, appears important in the development of diabetes-induced vascular dysfunction. Angiotensin-(1–7) [Ang-(1–7)] can prevent the development of hyperglycemia-induced vascular complications partly through inhibiting EGFR transactivation. Here, we investigated whether Ang-(1–7) can inhibit transactivation of ErbB2 as well as other ErbB receptors in vivo and in vitro. Streptozotocin-induced diabetic rats were chronically treated with Ang-(1–7) or AG825, a selective ErbB2 inhibitor, for 4 weeks and mechanistic studies performed in the isolated mesenteric vasculature bed as well as in cultured vascular smooth muscle cells (VSMCs). Ang-(1–7) or AG825 treatment inhibited diabetes-induced phosphorylation of ErbB2 receptor at tyrosine residues Y1221/22, Y1248, Y877, as well as downstream signaling via ERK1/2, p38 MAPK, ROCK, eNOS and IkB-α in the mesenteric vascular bed. In VSMCs cultured in high glucose (25 mM), Ang-(1–7) inhibited src-dependent ErbB2 transactivation that was opposed by the selective Mas receptor antagonist, D-Pro7-Ang-(1–7). Ang-(1–7) via Mas receptor also inhibited both Angiotensin II- and noradrenaline/norephinephrine-induced transactivation of ErbB2 and/or EGFR receptors. Further, hyperglycemia-induced transactivation of ErbB3 and ErbB4 receptors could be attenuated by Ang-(1–7) that could be prevented by D-Pro7-Ang-(1–7) in VSMC. These data suggest that Ang-(1–7) via its Mas receptor acts as a pan-ErbB inhibitor and might represent a novel general mechanism by which Ang-(1–7) exerts its beneficial effects in many disease states including diabetes-induced vascular complications.
Collapse
MESH Headings
- Angiotensin I/pharmacology
- Animals
- Blotting, Western
- Cells, Cultured
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/genetics
- Gene Expression Regulation/drug effects
- Glucose/metabolism
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Peptide Fragments/pharmacology
- Phosphorylation
- Proto-Oncogene Mas
- Proto-Oncogene Proteins/metabolism
- Rats
- Rats, Wistar
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction
- Transcriptional Activation/drug effects
- Vasodilator Agents/pharmacology
Collapse
Affiliation(s)
- Saghir Akhtar
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Safat 13110, Kuwait
- * E-mail:
| | - Bindu Chandrasekhar
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Safat 13110, Kuwait
| | - Sreeja Attur
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Safat 13110, Kuwait
| | - Gursev S. Dhaunsi
- Department of Pediatrics, Faculty of Medicine, Kuwait University, Safat 13110, Kuwait
| | - Mariam H. M. Yousif
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Safat 13110, Kuwait
| | - Ibrahim F. Benter
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Safat 13110, Kuwait
| |
Collapse
|
21
|
Benter IF, Sarkhou F, Al-Khaldi AT, Chandrasekhar B, Attur S, Dhaunsi GS, Yousif MHM, Akhtar S. The dual targeting of EGFR and ErbB2 with the inhibitor Lapatinib corrects high glucose-induced apoptosis and vascular dysfunction by opposing multiple diabetes-induced signaling changes. J Drug Target 2015; 23:506-18. [DOI: 10.3109/1061186x.2015.1057150] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
22
|
Abstract
PURPOSE OF THE REVIEW The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase with a wide implication in tumor biology, wound healing and development. Besides acting as a growth factor receptor activated by ligands such as EGF, the EGFR can also be transactivated and thereby mediate cross-talk with different signaling pathways. The aim of this review is to illustrate the Janus-faced function of the EGFR in the vasculature with its relevance for vascular biology and disease. RECENT FINDINGS Over recent years, the number of identified signaling partners of the EGFR has steadily increased, as have the biological processes in which the EGFR is thought to be involved. Recently, new models have allowed investigation of EGFR effects in vivo, shedding some light on the overall function of the EGFR in the vasculature. At the same time, EGFR inhibitors and antibodies have become increasingly established in cancer therapy, providing potential therapeutic tools for decreasing EGFR signaling. SUMMARY The EGFR is a versatile signaling pathway integrator associated with vascular homeostasis and disease. In addition to modulating basal vascular tone and tissue homeostasis, the EGFR also seems to be involved in proinflammatory, proliferative, migratory and remodeling processes, with enhanced deposition of extracellular matrix components, thereby promoting vascular diseases such as hypertension or atherosclerosis.
Collapse
|
23
|
The epidermal growth factor receptor and its ligands in cardiovascular disease. Int J Mol Sci 2013; 14:20597-613. [PMID: 24132149 PMCID: PMC3821633 DOI: 10.3390/ijms141020597] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 09/20/2013] [Accepted: 10/08/2013] [Indexed: 12/11/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) family and its ligands serve as a switchboard for the regulation of multiple cellular processes. While it is clear that EGFR activity is essential for normal cardiac development, its function in the vasculature and its role in cardiovascular disease are only beginning to be elucidated. In the blood vessel, endothelial cells and smooth muscle cells are both a source and a target of EGF-like ligands. Activation of EGFR has been implicated in blood pressure regulation, endothelial dysfunction, neointimal hyperplasia, atherogenesis, and cardiac remodeling. Furthermore, increased circulating EGF-like ligands may mediate accelerated vascular disease associated with chronic inflammation. Although EGFR inhibitors are currently being used clinically for the treatment of cancer, additional studies are necessary to determine whether abrogation of EGFR signaling is a potential strategy for the treatment of cardiovascular disease.
Collapse
|
24
|
Akhtar S, Yousif MHM, Dhaunsi GS, Sarkhouh F, Chandrasekhar B, Attur S, Benter IF. Activation of ErbB2 and Downstream Signalling via Rho Kinases and ERK1/2 Contributes to Diabetes-Induced Vascular Dysfunction. PLoS One 2013; 8:e67813. [PMID: 23826343 PMCID: PMC3694874 DOI: 10.1371/journal.pone.0067813] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Accepted: 05/21/2013] [Indexed: 12/20/2022] Open
Abstract
Diabetes mellitus leads to vascular complications but the underlying signalling mechanisms are not fully understood. Here, we examined the role of ErbB2 (HER2/Neu), a transmembrane receptor tyrosine kinase of the ErbB/EGFR (epidermal growth factor receptor) family, in mediating diabetes-induced vascular dysfunction in an experimental model of type 1 diabetes. Chronic treatment of streptozotocin-induced diabetic rats (1 mg/kg/alt diem) or acute, ex-vivo (10(-6), 10(-5) M) administration of AG825, a specific inhibitor of ErbB2, significantly corrected the diabetes-induced hyper-reactivity of the perfused mesenteric vascular bed (MVB) to the vasoconstrictor, norephinephrine (NE) and the attenuated responsiveness to the vasodilator, carbachol. Diabetes led to enhanced phosphorylation of ErbB2 at multiple tyrosine (Y) residues (Y1221/1222, Y1248 and Y877) in the MVB that could be attenuated by chronic AG825 treatment. Diabetes- or high glucose-mediated upregulation of ErbB2 phosphorylation was coupled with activation of Rho kinases (ROCKs) and ERK1/2 in MVB and in cultured vascular smooth muscle cells (VSMC) that were attenuated upon treatment with either chronic or acute AG825 or with anti-ErbB2 siRNA. ErbB2 likley heterodimerizes with EGFR, as evidenced by increased co-association in diabetic MVB, and further supported by our finding that ERK1/2 and ROCKs are common downstream effectors since their activation could also be blocked by AG1478. Our results show for the first time that ErbB2 is an upstream effector of ROCKs and ERK1/2 in mediating diabetes-induced vascular dysfunction. Thus, potential strategies aimed at modifying actions of signal transduction pathways involving ErbB2 pathway may prove to be beneficial in treatment of diabetes-induced vascular complications.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Diabetes Mellitus, Experimental/complications
- Diabetic Angiopathies/etiology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- MAP Kinase Signaling System
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Rats
- Rats, Wistar
- Receptor, ErbB-2/metabolism
- Transcriptional Activation
- Vasoconstrictor Agents/pharmacology
- Vasodilator Agents/pharmacology
- rho-Associated Kinases/metabolism
Collapse
Affiliation(s)
- Saghir Akhtar
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Safat, Kuwait
| | - Mariam H. M. Yousif
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Safat, Kuwait
| | - Gursev S. Dhaunsi
- Department of Pediatrics, Faculty of Medicine, Kuwait University, Safat, Kuwait
| | - Fatma Sarkhouh
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Safat, Kuwait
| | - Bindu Chandrasekhar
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Safat, Kuwait
| | - Sreeja Attur
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Safat, Kuwait
| | - Ibrahim F. Benter
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Safat, Kuwait
| |
Collapse
|
25
|
Combined Effects of PPAR γ Agonists and Epidermal Growth Factor Receptor Inhibitors in Human Proximal Tubule Cells. PPAR Res 2013; 2013:982462. [PMID: 23533381 PMCID: PMC3596915 DOI: 10.1155/2013/982462] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 01/25/2013] [Indexed: 12/28/2022] Open
Abstract
We aimed to determine whether epidermal growth factor receptor (EGFR) inhibition, in addition to a peroxisome proliferator-activated receptor gamma (PPARγ) agonist, prevents high-glucose-induced proximal tubular fibrosis, inflammation, and sodium and water retention in human proximal tubule cells exposed to normal glucose; high glucose; high glucose with the PPARγ agonist pioglitazone or with the P-EGFR inhibitor, gefitinib; or high glucose with both pioglitazone and gefitinib. We have shown that high glucose increases AP-1 and NFκB binding activity, downstream phosphorylation of EGFR and Erk1/2, and fibronectin and collagen IV expression. Pioglitazone reversed these effects but upregulated NHE3 and AQP1 expression. Gefitinib inhibited high glucose induced fibronectin and collagen IV, and EGFR and Erk1/2 phosphorylation and reversed pioglitazone-induced increases in NHE3 and AQP1 expression. Our data suggests that combination of an EGFR inhibitor and a PPARγ agonist mitigates high-glucose-induced fibrosis and inflammation and reverses the upregulation of transporters and channels involved in sodium and water retention in human proximal tubule cells. Hence EGFR blockade may hold promise, not only in limiting tubulointerstitial pathology in diabetic nephropathy, but also in limiting the sodium and water retention observed in patients with diabetes and exacerbated by PPARγ agonists.
Collapse
|
26
|
Benter IF, Babiker F, Al-Rashdan I, Yousif M, Akhtar S. RU28318, an aldosterone antagonist, in combination with an ACE inhibitor and angiotensin receptor blocker attenuates cardiac dysfunction in diabetes. J Diabetes Res 2013; 2013:427693. [PMID: 24066305 PMCID: PMC3771425 DOI: 10.1155/2013/427693] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 07/16/2013] [Accepted: 07/17/2013] [Indexed: 02/06/2023] Open
Abstract
AIMS We evaluated the effects of RU28318 (RU), a selective mineralocorticoid receptor (MR) antagonist, Captopril (Capt), an angiotensin converting enzyme inhibitor, and Losartan (Los), an angiotensin receptor blocker, alone or in combination with ischemia/reperfusion- (I/R-) induced cardiac dysfunction in hearts obtained from normal and diabetic rats. METHODS Isolated hearts were perfused for 30 min and then subjected to 30 min of global ischemia (I) followed by a period of 30 min of reperfusion (R). Drugs were administered for 30 min either before or after ischemia. Drug regimens tested were RU, Capt, Los, RU + Capt, RU + Los, Capt + Los, and RU + Capt + Los (Triple). Recovery of cardiac hemodynamics was evaluated. RESULTS Recovery of cardiac function was up to 5-fold worse in hearts obtained from diabetic animals compared to controls. Treatment with RU was generally better in preventing or reversing ischemia-induced cardiac dysfunction in normal hearts compared to treatment with Capt or Los alone. In diabetic hearts, RU was generally similarly effective as Capt or Los treatment. CONCLUSIONS RU treatment locally might be considered as an effective therapy or preventative measure in cardiac I/R injury. Importantly, RU was the most effective at improving -dP/dt (a measure of diastolic function) when administered to diabetic hearts after ischemia.
Collapse
Affiliation(s)
- Ibrahim F. Benter
- Department of Pharmacology & Toxicology, Faculty of Medicine, Kuwait University, P.O. Box 24923, 13110 Safat, Kuwait
| | - Fawzi Babiker
- Department of Physiology, Faculty of Medicine, Kuwait University, P.O. Box 24923, 13110 Safat, Kuwait
- *Fawzi Babiker:
| | - Ibrahim Al-Rashdan
- Department of Medicine, Faculty of Medicine, Kuwait University, P.O. Box 24923, 13110 Safat, Kuwait
| | - Mariam Yousif
- Department of Pharmacology & Toxicology, Faculty of Medicine, Kuwait University, P.O. Box 24923, 13110 Safat, Kuwait
| | - Saghir Akhtar
- Department of Pharmacology & Toxicology, Faculty of Medicine, Kuwait University, P.O. Box 24923, 13110 Safat, Kuwait
| |
Collapse
|
27
|
Akhtar S, Yousif MHM, Dhaunsi GS, Chandrasekhar B, Al-Farsi O, Benter IF. Angiotensin-(1-7) inhibits epidermal growth factor receptor transactivation via a Mas receptor-dependent pathway. Br J Pharmacol 2012; 165:1390-400. [PMID: 21806601 DOI: 10.1111/j.1476-5381.2011.01613.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE The transactivation of the epidermal growth factor (EGF) receptor appears to be an important central transduction mechanism in mediating diabetes-induced vascular dysfunction. Angiotensin-(1-7) [Ang-(1-7)] via its Mas receptor can prevent the development of hyperglycaemia-induced cardiovascular complications. Here, we investigated whether Ang-(1-7) can inhibit hyperglycaemia-induced EGF receptor transactivation and its classical signalling via ERK1/2 and p38 MAPK in vivo and in vitro. EXPERIMENTAL APPROACH Streptozotocin-induced diabetic rats were chronically treated with Ang-(1-7) or AG1478, a selective EGF receptor inhibitor, for 4 weeks and mechanistic studies performed in the isolated mesenteric vasculature bed as well as in primary cultures of vascular smooth muscle cells (VSMCs). KEY RESULTS Diabetes significantly enhanced phosphorylation of EGF receptor at tyrosine residues Y992, Y1068, Y1086, Y1148, as well as ERK1/2 and p38 MAPK in the mesenteric vasculature bed whereas these changes were significantly attenuated upon Ang-(1-7) or AG1478 treatment. In VSMCs grown in conditions of high glucose (25 mM), an Src-dependent elevation in EGF receptor phosphorylation was observed. Ang-(1-7) inhibited both Ang II- and glucose-induced transactivation of EGF receptor. The inhibition of high glucose-mediated Src-dependant transactivation of EGF receptor by Ang-(1-7) could be prevented by a selective Mas receptor antagonist, D-Pro7-Ang-(1-7). CONCLUSIONS AND IMPLICATIONS These results show for the first time that Ang-(1-7) inhibits EGF receptor transactivation via a Mas receptor/Src-dependent pathway and might represent a novel general mechanism by which Ang-(1-7) exerts its beneficial effects in many disease states including diabetes-induced vascular dysfunction.
Collapse
Affiliation(s)
- Saghir Akhtar
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Safat, Kuwait.
| | | | | | | | | | | |
Collapse
|
28
|
Panchapakesan U, Pollock C, Saad S. Renal epidermal growth factor receptor: its role in sodium and water homeostasis in diabetic nephropathy. Clin Exp Pharmacol Physiol 2011; 38:84-8. [PMID: 21155863 DOI: 10.1111/j.1440-1681.2010.05472.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
1. Volume expansion is observed in animal and human models of diabetic nephropathy, which is in a large part a result of disordered renal tubular cell sodium and water transport. 2. Sodium transport in the proximal tubule is increased in diabetes mellitus as a result of enhanced activity of the sodium-hydrogen exchanger-3 (NHE3), the key transporter for transcellular reabsorption of sodium. Transactivation of the epidermal growth factor receptor (EGFR) by factors inherent in the milieu of diabetes mellitus increases serum glucocorticoid regulated kinase-1 (Sgk1), a key regulator of NHE3. 3. Enhanced sodium and water reabsorption, occurring as a consequence of endogenous or pharmacological stimulation of the peroxisome proliferator-activated receptor gamma is Sgk1 mediated. 4. EGFR inhibitors, which are currently used clinically to treat malignancies, might have potential in attenuating the cellular mechanisms responsible for thiazolidinedione (TZD)-mediated sodium and water transport in diabetes. 5. In the present review, the authors focus on the importance of the EGFR in sodium and water uptake in the proximal tubule in the environment of pathophysiological and pharmacological influences.
Collapse
Affiliation(s)
- Usha Panchapakesan
- Renal Research Group, Department of Medicine, University of Sydney, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | | | | |
Collapse
|
29
|
Matrougui K. Diabetes and microvascular pathophysiology: role of epidermal growth factor receptor tyrosine kinase. Diabetes Metab Res Rev 2010; 26:13-6. [PMID: 19943320 PMCID: PMC2823570 DOI: 10.1002/dmrr.1050] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Type 2 diabetes is responsible for the increased prevalence of ischaemic heart disease, generally related to coronary artery disease, which is associated with increased morbidity and death in diabetic patients. Epidermal growth factor receptor (EGFR) tyrosine kinase, one of the many factors involved in cell growth and migration, has been shown to be key element in the development of microvessel myogenic tone. In a recent study, we have shown that microvascular dysfunction in type 2 diabetes is dependent on the exacerbation of the EGFR tyrosine kinase phosphorylation. Thus, further elucidation of this EGFR transactivation and down stream signalling will offer a new direction to investigate the mechanism of microvascular dysfunction responsible for heart disease that occurs in type 2 diabetes. In this review, we discuss the link between the EGFR transactivation and microvascular dysfunction that occurs in type 2 diabetes.
Collapse
Affiliation(s)
- Khalid Matrougui
- Department of Physiology, and Hypertension and Renal Center of Excellence, Tulane University, 1430 Tulane Ave., New Orleans, LA 70112, USA.
| |
Collapse
|