1
|
Ramasamy A, Mohan C. Molecular and Cellular Mediators of Renal Fibrosis in Lupus Nephritis. Int J Mol Sci 2025; 26:2621. [PMID: 40141260 PMCID: PMC11942537 DOI: 10.3390/ijms26062621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/28/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Lupus nephritis (LN), a significant complication of systemic lupus erythematosus (SLE), represents a challenging manifestation of the disease. One of the prominent pathophysiologic mechanisms targeting the renal parenchyma is fibrosis, a terminal process resulting in irreversible tissue damage that eventually leads to a decline in renal function and/or end-stage kidney disease (ESKD). Both glomerulosclerosis and interstitial fibrosis emerge as reliable prognostic indicators of renal outcomes. This article reviews the hallmarks of renal fibrosis in lupus nephritis, including the known and putative drivers of fibrogenesis. A better understanding of the cellular and molecular processes driving fibrosis in LN may help inform the development of therapeutic strategies for this disease, as well as the identification of individuals at higher risk of developing ESKD.
Collapse
Affiliation(s)
| | - Chandra Mohan
- Biomedical Engineering Department, University of Houston, 3517 Cullen Blvd, Room 2027, Houston, TX 77204, USA;
| |
Collapse
|
2
|
Kono DH, Hahn BH. Animal models of systemic lupus erythematosus (SLE). DUBOIS' LUPUS ERYTHEMATOSUS AND RELATED SYNDROMES 2025:189-234. [DOI: 10.1016/b978-0-323-93232-5.00024-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
3
|
Méndez-García LA, Escobedo G, Baltazar-Pérez I, Ocampo-Aguilera NA, Arreola-Miranda JA, Cid-Soto MA, Alfaro-Cruz A, González-Chávez A, Ocaña-Guzmán AR, Solleiro-Villavicencio H. Exploring the Th2 Response in Obesity and Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): A Potential Modulator of the Renin-Angiotensin System (RAS) Pathway in Hypertension Development. Life (Basel) 2024; 14:1080. [PMID: 39337863 PMCID: PMC11433558 DOI: 10.3390/life14091080] [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: 08/09/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), now referred to as metabolic dysfunction-associated steatotic liver disease (MASLD), is alarmingly increasing alongside the cases of obesity worldwide. MASLD is an underestimated metabolic abnormality closely linked with a higher risk of developing systemic arterial hypertension (SAH). However, the underlying mechanism of association between MASLD and SAH remains unknown. Inflammation may link these two entities by regulating the renin-angiotensin system (RAS). For this reason, in this study, we evaluated the hepatic expression of a cytokine profile and critical molecules in the RAS pathway in patients with morbid obesity and MASLD, both with SAH. We found a statistically significant correlation between ACE levels and the cytokines IL-4, IL-10, and IL-13 of Th2 response. Furthermore, according to a multiple linear regression analysis, the cytokines IL-4 and IL-13 were the best predictors of ACE levels. Moreover, we observed increased hepatic IL-13 expression in patients with morbid obesity, MASLD, and SAH compared to those without SAH. These results allow us to propose, for the first time, that the Th2 response, through regulating the RAS, could play a critical role in developing SAH in individuals with MASLD and obesity.
Collapse
Affiliation(s)
- Lucía Angélica Méndez-García
- Immunometabolism Laboratory, General Hospital of Mexico “Eduardo Liceaga”, Mexico City 06720, Mexico; (G.E.); (I.B.-P.); (N.A.O.-A.); (J.A.A.-M.)
| | - Galileo Escobedo
- Immunometabolism Laboratory, General Hospital of Mexico “Eduardo Liceaga”, Mexico City 06720, Mexico; (G.E.); (I.B.-P.); (N.A.O.-A.); (J.A.A.-M.)
| | - Itzel Baltazar-Pérez
- Immunometabolism Laboratory, General Hospital of Mexico “Eduardo Liceaga”, Mexico City 06720, Mexico; (G.E.); (I.B.-P.); (N.A.O.-A.); (J.A.A.-M.)
- Genomics Sciences Program, Oncogenomics and Cancer Proteomics Laboratory, Autonomous University of Mexico City, Avenue San Lorenzo 290, Mexico City 03100, Mexico
| | - Nydia Angélica Ocampo-Aguilera
- Immunometabolism Laboratory, General Hospital of Mexico “Eduardo Liceaga”, Mexico City 06720, Mexico; (G.E.); (I.B.-P.); (N.A.O.-A.); (J.A.A.-M.)
- Genomics Sciences Program, Oncogenomics and Cancer Proteomics Laboratory, Autonomous University of Mexico City, Avenue San Lorenzo 290, Mexico City 03100, Mexico
| | - José Alfonso Arreola-Miranda
- Immunometabolism Laboratory, General Hospital of Mexico “Eduardo Liceaga”, Mexico City 06720, Mexico; (G.E.); (I.B.-P.); (N.A.O.-A.); (J.A.A.-M.)
- Genomics Sciences Program, Oncogenomics and Cancer Proteomics Laboratory, Autonomous University of Mexico City, Avenue San Lorenzo 290, Mexico City 03100, Mexico
| | - Miguel Angel Cid-Soto
- Sequencing Laboratory, Division of Research Development, National Medical Center “Siglo XXI”, Mexican Social Security Institute, Mexico City 06720, Mexico;
| | - Ana Alfaro-Cruz
- Pathological Anatomy Department, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico;
| | - Antonio González-Chávez
- Comprehensive Care Clinic for Patients with Diabetes and Obesity (CAIDO), General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico;
| | | | - Helena Solleiro-Villavicencio
- Genomics Sciences Program, Oncogenomics and Cancer Proteomics Laboratory, Autonomous University of Mexico City, Avenue San Lorenzo 290, Mexico City 03100, Mexico
| |
Collapse
|
4
|
Cieślik M, Strobel SD, Bryniarski P, Twardowska H, Chmielowski A, Rudek M, Felkle D, Zięba K, Kaleta K, Jarczyński M, Nowak B, Bryniarski K, Nazimek K. Hypotensive drugs mitigate the high-sodium diet-induced pro-inflammatory activation of mouse macrophages in vivo. Biomed Pharmacother 2024; 175:116648. [PMID: 38677242 DOI: 10.1016/j.biopha.2024.116648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024] Open
Abstract
Nowadays, there is an increasing emphasis on the need to alleviate the chronic inflammatory response to effectively treat hypertension. However, there are still gaps in our understanding on how to achieve this. Therefore, research on interaction of antihypertensive drugs with the immune system is extremely interesting, since their therapeutic effect could partly result from amelioration of hypertension-related inflammation, in which macrophages seem to play a pivotal role. Thus, current comprehensive studies have investigated the impact of repeatedly administered hypotensive drugs (captopril, olmesartan, propranolol, carvedilol, amlodipine, verapamil) on macrophage functions in the innate and adaptive immunity, as well as if drug-induced effects are affected by a high-sodium diet (HSD), one of the key environmental risk factors of hypertension. Although the assayed medications increased the generation of reactive oxygen and nitrogen intermediates by macrophages from standard fed donors, they reversed HSD-induced enhancing effects on macrophage oxidative burst and secretion of pro-inflammatory cytokines. On the other hand, some drugs increased macrophage phagocytic activity and the expression of surface markers involved in antigen presentation, which translated into enhanced macrophage ability to activate B cells for antibody production. Moreover, the assayed medications augmented macrophage function and the effector phase of contact hypersensitivity reaction, but suppressed the sensitization phase of cell-mediated hypersensitivity under HSD conditions. Our current findings contribute to the recognition of mechanisms, by which excessive sodium intake affects macrophage immune activity in hypertensive individuals, and provide evidence that the assayed medications mitigate most of the HSD-induced adverse effects, suggesting their additional protective therapeutic activity.
Collapse
Affiliation(s)
- Martyna Cieślik
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Spencer D Strobel
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Paweł Bryniarski
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Hanna Twardowska
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Adam Chmielowski
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Michał Rudek
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Dominik Felkle
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Katarzyna Zięba
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Konrad Kaleta
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Mateusz Jarczyński
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Bernadeta Nowak
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Krzysztof Bryniarski
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Katarzyna Nazimek
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland.
| |
Collapse
|
5
|
Miñano S, González-Correa C, Moleón J, Duarte J. Metabolic Modulators in Cardiovascular Complications of Systemic Lupus Erythematosus. Biomedicines 2023; 11:3142. [PMID: 38137363 PMCID: PMC10741086 DOI: 10.3390/biomedicines11123142] [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: 10/20/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifactorial disorder with contributions from hormones, genetics, and the environment, predominantly affecting young women. Cardiovascular disease is the primary cause of mortality in SLE, and hypertension is more prevalent among SLE patients. The dysregulation of both innate and adaptive immune cells in SLE, along with their infiltration into kidney and vascular tissues, is a pivotal factor contributing to the cardiovascular complications associated with SLE. The activation, proliferation, and differentiation of CD4+ T cells are intricately governed by cellular metabolism. Numerous metabolic inhibitors have been identified to target critical nodes in T cell metabolism. This review explores the existing evidence and knowledge gaps concerning whether the beneficial effects of metabolic modulators on autoimmunity, hypertension, endothelial dysfunction, and renal injury in lupus result from the restoration of a balanced immune system. The inhibition of glycolysis, mitochondrial metabolism, or mTORC1 has been found to improve endothelial dysfunction and prevent the development of hypertension in mouse models of SLE. Nevertheless, limited information is available regarding the potential vasculo-protective effects of drugs that act on immunometabolism in SLE patients.
Collapse
Affiliation(s)
- Sofía Miñano
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (S.M.); (C.G.-C.)
| | - Cristina González-Correa
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (S.M.); (C.G.-C.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
| | - Javier Moleón
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (S.M.); (C.G.-C.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (S.M.); (C.G.-C.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| |
Collapse
|
6
|
Felkle D, Zięba K, Kaleta K, Czaja J, Zyzdorf A, Sobocińska W, Jarczyński M, Bryniarski K, Nazimek K. Overreactive macrophages in SARS-CoV-2 infection: The effects of ACEI. Int Immunopharmacol 2023; 124:110858. [PMID: 37708705 DOI: 10.1016/j.intimp.2023.110858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/16/2023]
Abstract
Among various factors influencing the course of SARS-CoV-2 infection in humans, macrophage overactivation is considered the main cause of the cytokine storm that leads to severe complications of COVID-19. Moreover, the increased expression of angiotensin converting enzyme 2 (ACE2), an obligatory entry receptor of the coronavirus, caused by treatment with ACE inhibitors (ACEI) lowered overall confidence in the safety of these drugs. However, analysis of the course of coronavirus infection in patients treated with ACEI does not support these concerns. Instead, the beneficial effect of ACEI on macrophages has increasingly been emphasized. This includes their anti-inflammatory activation and the consequent reduction in the risk of severe disease and life-threatening complications. Herein, we summarize the current knowledge and understanding of the dual role of macrophages in SARS-CoV-2 infection, with a special focus on the postulated mechanisms underlying the beneficial effects of macrophage targeting by ACEI. These seem to involve the stimulation of macrophage angiotensin II type 2 and Mas receptors by angiotensin 1-7, intensively produced due to the up-regulation of ACE2 expression on macrophages, as well as the direct inhibition of macrophage hyper-responsiveness by ACEI. The impact of ACEI on macrophages may also lead to the activation of an effective antiviral response due to the increased expression of ACE2.
Collapse
Affiliation(s)
- Dominik Felkle
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Katarzyna Zięba
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Konrad Kaleta
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Julia Czaja
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Amanda Zyzdorf
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Wiktoria Sobocińska
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Mateusz Jarczyński
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Krzysztof Bryniarski
- Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Katarzyna Nazimek
- Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland.
| |
Collapse
|
7
|
Copur S, Peltek IB, Mutlu A, Tanriover C, Kanbay M. A new immune disease: systemic hypertension. Clin Kidney J 2023; 16:1403-1419. [PMID: 37664577 PMCID: PMC10469084 DOI: 10.1093/ckj/sfad059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Indexed: 09/05/2023] Open
Abstract
Systemic hypertension is the most common medical comorbidity affecting the adult population globally, with multiple associated outcomes including cerebrovascular diseases, cardiovascular diseases, vascular calcification, chronic kidney disease, metabolic syndrome and mortality. Despite advancements in the therapeutic field approximately one in every five adult patients with hypertension is classified as having treatment-resistant hypertension, indicating the need for studies to provide better understanding of the underlying pathophysiology and the need for more therapeutic targets. Recent pre-clinical studies have demonstrated the role of the innate and adaptive immune system including various cell types and cytokines in the pathophysiology of hypertension. Moreover, pre-clinical studies have indicated the potential beneficial effects of immunosuppressant medications in the control of hypertension. Nevertheless, it is unclear whether such pathophysiological mechanisms and therapeutic alternatives are applicable to human subjects, while this area of research is undoubtedly a rapidly growing field.
Collapse
Affiliation(s)
- Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Ibrahim B Peltek
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Ali Mutlu
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Cem Tanriover
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Mehmet Kanbay
- Department of Medicine, Section of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| |
Collapse
|
8
|
Xia Y, Jiang C, Yang M, Liu T, Zou X, Li C, Wang X. SB431542 alleviates lupus nephritis by regulating B cells and inhibiting the TLR9/TGFβ1/PDGFB signaling. J Autoimmun 2022; 132:102894. [PMID: 36030617 DOI: 10.1016/j.jaut.2022.102894] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/09/2022] [Accepted: 08/09/2022] [Indexed: 11/28/2022]
Abstract
Lupus nephritis (LN) is the most common cause of morbidity and mortality in patients with systemic lupus erythematosus (SLE). Currently, immunosuppressive treatments for LN are suboptimal and can induce significant side effects. SB431542 is a selective and potent inhibitor of the TGFβ/Activin/NODAL pathway. Here, we study the effects of SB431542 treatment on LN and discuss the potential mechanisms. SB431542 ameliorated clinical outcomes with a consequent histological improvement in NZB/W mice. A comparative transcriptional profiling analysis revealed 586 differentially expressed genes (247 downregulated genes) in the SB431542 group compared to the control group. We found that the downregulated genes were mainly enriched in the biological processes of B cell activation, B cell proliferation, B cell differentiation, and B cell receptor signaling. Kyoto encyclopedia of genes and genomes pathway analysis revealed that the hematopoietic cell linage pathway was significantly downregulated in the SB431542 group. In addition, we observed that SB431542 reduced the splenic or renal levels of CD20 and the serum levels of anti-dsDNA antibody (IgG) in NZB/W mice. Furthermore, qRT-PCR and immunohistochemistry confirmed that SB431542 inhibits the production of TLR9, TGFβ1, and PDGFB. Thus, due to its immunomodulatory activities, SB431542 could be considered for clinical therapy development for LN.
Collapse
Affiliation(s)
- Ying Xia
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Chuan Jiang
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Mingyue Yang
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Tao Liu
- Department of Rheumatology and Immunology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xiaojuan Zou
- Department of Rheumatology and Immunology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Chenxu Li
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xiaosong Wang
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
9
|
Exploring the Impact of ACE Inhibition in Immunity and Disease. J Renin Angiotensin Aldosterone Syst 2022; 2022:9028969. [PMID: 36016727 PMCID: PMC9371878 DOI: 10.1155/2022/9028969] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/07/2022] [Indexed: 11/18/2022] Open
Abstract
Angiotensin-converting enzyme (ACE) is a zinc-dependent dipeptidyl carboxypeptidase and is crucial in the renin-angiotensin-aldosterone system (RAAS) but also implicated in immune regulation. Intrinsic ACE has been detected in several immune cell populations, including macrophages and neutrophils, where its overexpression results in enhanced bactericidal and antitumour responses, independent of angiotensin II. With roles in antigen presentation and inflammation, the impact of ACE inhibitors must be explored to understand how ACE inhibition may impact our ability to clear infections or malignancy, particularly in the wake of the coronavirus (SARS-CoV2) pandemic and as antibiotic resistance grows. Patients using ACE inhibitors may be more at risk of postsurgical complications as ACE inhibition in human neutrophils results in decreased ROS and phagocytosis whilst angiotensin receptor blockers (ARBs) have no effect. In contrast, ACE is also elevated in certain autoimmune diseases such as rheumatoid arthritis and lupus, and its inhibition benefits patient outcome where inflammatory immune cells are overactive. Although the ACE autoimmune landscape is changing, some studies have conflicting results and require further input. This review seeks to highlight the need for further research covering ACE inhibitor therapeutics and their potential role in improving autoimmune conditions, cancer, or how they may contribute to immunocompromise during infection and neurodegenerative diseases. Understanding ACE inhibition in immune cells is a developing field that will alter how ACE inhibitors are designed in future and aid in developing therapeutic interventions.
Collapse
|
10
|
Cunha PS, Laranjo S, Heijman J, Oliveira MM. The Atrium in Atrial Fibrillation - A Clinical Review on How to Manage Atrial Fibrotic Substrates. Front Cardiovasc Med 2022; 9:879984. [PMID: 35859594 PMCID: PMC9289204 DOI: 10.3389/fcvm.2022.879984] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 06/03/2022] [Indexed: 12/27/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia in the population and is associated with a significant clinical and economic burden. Rigorous assessment of the presence and degree of an atrial arrhythmic substrate is essential for determining treatment options, predicting long-term success after catheter ablation, and as a substrate critical in the pathophysiology of atrial thrombogenesis. Catheter ablation of AF has developed into an essential rhythm-control strategy. Nowadays is one of the most common cardiac ablation procedures performed worldwide, with its success inversely related to the extent of atrial structural disease. Although atrial substrate evaluation remains complex, several diagnostic resources allow for a more comprehensive assessment and quantification of the extent of left atrial structural remodeling and the presence of atrial fibrosis. In this review, we summarize the current knowledge on the pathophysiology, etiology, and electrophysiological aspects of atrial substrates promoting the development of AF. We also describe the risk factors for its development and how to diagnose its presence using imaging, electrocardiograms, and electroanatomic voltage mapping. Finally, we discuss recent data regarding fibrosis biomarkers that could help diagnose atrial fibrotic substrates.
Collapse
Affiliation(s)
- Pedro Silva Cunha
- Arrhythmology, Pacing and Electrophysiology Unit, Cardiology Service, Santa Marta Hospital, Central Lisbon Hospital University Center, Lisbon, Portugal
- Lisbon School of Medicine, Universidade de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Center, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Sérgio Laranjo
- Arrhythmology, Pacing and Electrophysiology Unit, Cardiology Service, Santa Marta Hospital, Central Lisbon Hospital University Center, Lisbon, Portugal
- Lisbon School of Medicine, Universidade de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Center, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Mário Martins Oliveira
- Arrhythmology, Pacing and Electrophysiology Unit, Cardiology Service, Santa Marta Hospital, Central Lisbon Hospital University Center, Lisbon, Portugal
- Lisbon School of Medicine, Universidade de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Center, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
11
|
The immunomodulatory effects of antihypertensive therapy: A review. Biomed Pharmacother 2022; 153:113287. [PMID: 35728352 DOI: 10.1016/j.biopha.2022.113287] [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: 04/26/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/23/2022] Open
Abstract
Hypertension remains the leading preventable risk factor for stroke and coronary artery disease, significantly contributing to all-cause global mortality and predisposing patients to renal and heart failure, as well as peripheral vascular disease. Due to the widespread usage of antihypertensive drugs, global mean blood pressure has remained unchanged or even slightly decreased over the past four decades. However, considering the broad spectrum of mechanisms involved in the action of antihypertensive drugs and the prevalence of their target receptors on immune cells, possible immunomodulatory effects which may exert beneficial effects of lowering blood pressure but also potentially alter immune function should be considered. In this review, we attempt to assess the consequences to immune system function of administering the five most commonly prescribed groups of antihypertensive drugs and to explain the mechanisms behind those interactions. Finally, we show potential gaps in our understanding of the effects of antihypertensive drugs on patient health. With regard to the widespread use of these drugs in the adult population worldwide, the discussed results may be of vital importance to evidence-based decision-making in daily clinical practice.
Collapse
|
12
|
Chaudhari S, Pham GS, Brooks CD, Dinh VQ, Young-Stubbs CM, Shimoura CG, Mathis KW. Should Renal Inflammation Be Targeted While Treating Hypertension? Front Physiol 2022; 13:886779. [PMID: 35770194 PMCID: PMC9236225 DOI: 10.3389/fphys.2022.886779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/17/2022] [Indexed: 11/28/2022] Open
Abstract
Despite extensive research and a plethora of therapeutic options, hypertension continues to be a global burden. Understanding of the pathological roles of known and underexplored cellular and molecular pathways in the development and maintenance of hypertension is critical to advance the field. Immune system overactivation and inflammation in the kidneys are proposed alternative mechanisms of hypertension, and resistant hypertension. Consideration of the pathophysiology of hypertension in chronic inflammatory conditions such as autoimmune diseases, in which patients present with autoimmune-mediated kidney inflammation as well as hypertension, may reveal possible contributors and novel therapeutic targets. In this review, we 1) summarize current therapies used to control blood pressure and their known effects on inflammation; 2) provide evidence on the need to target renal inflammation, specifically, and especially when first-line and combinatory treatment efforts fail; and 3) discuss the efficacy of therapies used to treat autoimmune diseases with a hypertension/renal component. We aim to elucidate the potential of targeting renal inflammation in certain subsets of patients resistant to current therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Keisa W. Mathis
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
13
|
Boesen EI, Kakalij RM. Autoimmune-mediated renal disease and hypertension. Clin Sci (Lond) 2021; 135:2165-2196. [PMID: 34533582 PMCID: PMC8477620 DOI: 10.1042/cs20200955] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 08/20/2021] [Accepted: 09/06/2021] [Indexed: 12/18/2022]
Abstract
Hypertension is a major risk factor for cardiovascular disease, chronic kidney disease (CKD), and mortality. Troublingly, hypertension is highly prevalent in patients with autoimmune renal disease and hastens renal functional decline. Although progress has been made over the past two decades in understanding the inflammatory contributions to essential hypertension more broadly, the mechanisms active in autoimmune-mediated renal diseases remain grossly understudied. This Review provides an overview of the pathogenesis of each of the major autoimmune diseases affecting the kidney that are associated with hypertension, and describes the current state of knowledge regarding hypertension in these diseases and their management. Specifically, discussion focuses on Systemic Lupus Erythematosus (SLE) and Lupus Nephritis (LN), Immunoglobulin A (IgA) Nephropathy, Idiopathic Membranous Nephropathy (IMN), Anti-Neutrophil Cytoplasmic Antibody (ANCA)-associated glomerulonephritis, and Thrombotic Thrombocytopenic Purpura (TTP). A summary of disease-specific animal models found to exhibit hypertension is also included to highlight opportunities for much needed further investigation of underlying mechanisms and novel therapeutic approaches.
Collapse
Affiliation(s)
- Erika I Boesen
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, U.S.A
| | - Rahul M Kakalij
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, U.S.A
| |
Collapse
|
14
|
Simões E Silva AC, Lanza K, Palmeira VA, Costa LB, Flynn JT. 2020 update on the renin-angiotensin-aldosterone system in pediatric kidney disease and its interactions with coronavirus. Pediatr Nephrol 2021; 36:1407-1426. [PMID: 32995920 PMCID: PMC7524035 DOI: 10.1007/s00467-020-04759-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/12/2020] [Accepted: 09/03/2020] [Indexed: 12/16/2022]
Abstract
The last decade was crucial for our understanding of the renin-angiotensin-aldosterone system (RAAS) as a two-axis, counter-regulatory system, divided into the classical axis, formed by angiotensin-converting enzyme (ACE), angiotensin II (Ang II), and the angiotensin type 1 receptor (AT1R), and the alternative axis comprising angiotensin-converting enzyme 2 (ACE2), angiotensin-(1-7) (Ang-(1-7)), and the Mas receptor. Breakthrough discoveries also took place, with other RAAS endopeptides being described, including alamandine and angiotensin A. In this review, we characterize the two RAAS axes and the role of their components in pediatric kidney diseases, including childhood hypertension (HTN), pediatric glomerular diseases, congenital abnormalities of the kidney and urinary tract (CAKUT), and chronic kidney disease (CKD). We also present recent findings on potential interactions between the novel coronavirus, SARS-CoV-2, and components of the RAAS, as well as potential implications of coronavirus disease 2019 (COVID-19) for pediatric kidney diseases.
Collapse
Affiliation(s)
- Ana Cristina Simões E Silva
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Avenida Alfredo Balena, 190, 2nd floor, Room # 281, Belo Horizonte, MG, 30130-100, Brazil.
- Pediatric Nephrology Unit, Department of Pediatrics, Faculty of Medicine, UFMG, Belo Horizonte, Brazil.
| | - Katharina Lanza
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Avenida Alfredo Balena, 190, 2nd floor, Room # 281, Belo Horizonte, MG, 30130-100, Brazil
| | - Vitória Andrade Palmeira
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Avenida Alfredo Balena, 190, 2nd floor, Room # 281, Belo Horizonte, MG, 30130-100, Brazil
| | - Larissa Braga Costa
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Avenida Alfredo Balena, 190, 2nd floor, Room # 281, Belo Horizonte, MG, 30130-100, Brazil
| | - Joseph T Flynn
- Pediatric Nephrology, Seattle Children's Hospital, University of Washington School of Medicine, Seattle, WA, 98105, USA
| |
Collapse
|
15
|
Nocito C, Lubinsky C, Hand M, Khan S, Patel T, Seliga A, Winfield M, Zuluaga-Ramirez V, Fernandes N, Shi X, Unterwald EM, Persidsky Y, Sriram U. Centrally Acting Angiotensin-Converting Enzyme Inhibitor Suppresses Type I Interferon Responses and Decreases Inflammation in the Periphery and the CNS in Lupus-Prone Mice. Front Immunol 2020; 11:573677. [PMID: 33042154 PMCID: PMC7522287 DOI: 10.3389/fimmu.2020.573677] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by multi-organ damage. Neuropsychiatric lupus (NPSLE) is one of the most common manifestations of human SLE, often causing depression. Interferon-α (IFNα) is a central mediator in disease pathogenesis. Administration of IFNα to patients with chronic viral infections or cancers causes depressive symptoms. Angiotensin-converting enzyme (ACE) is part of the kallikrein-kinin/renin-angiotensin (KKS/RAS) system that regulates many physiological processes, including inflammation, and brain functions. It is known that ACE degrades bradykinin (BK) into inactive peptides. We have previously shown in an in vitro model of mouse bone-marrow-derived dendritic cells (BMDC) and human peripheral blood mononuclear cells that captopril (a centrally acting ACE inhibitor-ACEi) suppressed Type I IFN responsive gene (IRG) expression. In this report, we used the MRL/lpr lupus-prone mouse model, an established model to study NPSLE, to determine the in vivo effects of captopril on Type I IFN and associated immune responses in the periphery and brain and effects on behavior. Administering captopril to MRL/lpr mice decreased expression of IRGs in brain, spleen and kidney, decreased circulating and tissue IFNα levels, decreased microglial activation (IBA-1 expression) and reduced depressive-like behavior. Serotonin levels that are decreased in depression were increased by captopril treatment. Captopril also reduced autoantibody levels in plasma and immune complex deposition in kidney and brain. Thus, ACEi's may have potential for therapeutic use for systemic and NPSLE.
Collapse
Affiliation(s)
- Cassandra Nocito
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Cody Lubinsky
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Michelle Hand
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Sabeeya Khan
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Tulsi Patel
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Alecia Seliga
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Malika Winfield
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Viviana Zuluaga-Ramirez
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Nicole Fernandes
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Xiangdang Shi
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Ellen M Unterwald
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Yuri Persidsky
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Uma Sriram
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
16
|
Ambari AM, Setianto B, Santoso A, Radi B, Dwiputra B, Susilowati E, Tulrahmi F, Doevendans PA, Cramer MJ. Angiotensin Converting Enzyme Inhibitors (ACEIs) Decrease the Progression of Cardiac Fibrosis in Rheumatic Heart Disease Through the Inhibition of IL-33/sST2. Front Cardiovasc Med 2020; 7:115. [PMID: 32850979 PMCID: PMC7399157 DOI: 10.3389/fcvm.2020.00115] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022] Open
Abstract
Rheumatic heart disease (RHD) is common in developing countries and poses a big medical challenge and burden. The pathogenesis of RHD is influenced by the triad of host, agent, and environment. Autoantigens generated from Group A Streptococcus (GAS) infection are captured by the resident dendritic cells (DCs) in the heart's valvular endothelium. DCs differentiate into antigen presenting cells (APC) in the valve interstices. APC induces activation of autoreactive T cells, which triggers inflammation and tissue fibrosis. Cardiac fibrosis is promoted through the activation of Mitogen activated protein kinases (MAPKs) and its downstream signaling, including its interaction with transforming growth factor-β (TGF-β) and Smad proteins. TGF-β-induced phosphorylation of Smad2 complexes with Smad3 and Smad4, and translocates into the nucleus. Angiotensin II enhances the migration, maturation, and presentation of DC. In RHD, Angiotensin II induces fibrosis via the stimulation of TGF-β, which further increases the binding of IL-33 to sST2 but not ST2L, resulting in the upregulation of Angiotensin II and progression of cardiac fibrosis. This cascade of inflammation and valvular fibrosis causes calcification and stiffening of the heart valves in RHD. Angiotensin converting enzyme inhibitors (ACEIs) inhibit Angiotensin II production, which in turn decreases TGF-β expression and the onset of overt inflammatory response. This condition leads to a reduction in the sST2 as the decoy receptor to "steal" IL-33, and IL-33 binds to ST2L and results in cardioprotection against cardiac fibrosis in the pathogenesis of RHD.
Collapse
Affiliation(s)
- Ade M. Ambari
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, National Cardiovascular Center Harapan Kita, University of Indonesia, Jakarta, Indonesia
| | - Budhi Setianto
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, National Cardiovascular Center Harapan Kita, University of Indonesia, Jakarta, Indonesia
| | - Anwar Santoso
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, National Cardiovascular Center Harapan Kita, University of Indonesia, Jakarta, Indonesia
| | - Basuni Radi
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, National Cardiovascular Center Harapan Kita, University of Indonesia, Jakarta, Indonesia
| | - Bambang Dwiputra
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, National Cardiovascular Center Harapan Kita, University of Indonesia, Jakarta, Indonesia
| | - Eliana Susilowati
- Research Assistants of Preventive Cardiology, National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Fadilla Tulrahmi
- Research Assistants of Preventive Cardiology, National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Pieter A. Doevendans
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
- Cardiovascular Departement, The Netherlands Heart Institute Utrecht, Utrecht, Netherlands
| | - Maarten J. Cramer
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
17
|
Munguia-Realpozo P, Mendoza-Pinto C, Sierra Benito C, Escarcega RO, Garcia-Carrasco M, Mendez Martinez S, Etchegaray Morales I, Galvez Romero JL, Ruiz-Arguelles A, Cervera R. Systemic lupus erythematosus and hypertension. Autoimmun Rev 2019; 18:102371. [DOI: 10.1016/j.autrev.2019.102371] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 04/02/2019] [Indexed: 01/01/2023]
|
18
|
Almaani S, Parikh SV. Membranous Lupus Nephritis: A Clinical Review. Adv Chronic Kidney Dis 2019; 26:393-403. [PMID: 31733724 DOI: 10.1053/j.ackd.2019.08.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/01/2019] [Accepted: 08/19/2019] [Indexed: 01/09/2023]
Abstract
Membranous lupus nephritis (MLN) (Class V lupus nephritis [LN]) is a distinct form of LN defined by the presence of subepithelial immune complex deposits seen on kidney biopsy. MLN is often associated with the nephrotic syndrome. The histology of MLN closely resembles that of idiopathic (primary) membranous nephropathy (pMN). However, MLN typically has abundant mesangial deposits that are absent in primary membranous nephropathy. The clinical presentation, management, and prognosis of MLN differ from that of the proliferative forms of LN (Class III, IV, or Mixed III/IV + V). Although immunosuppressive therapy is often warranted in MLN, the optimal treatment regimen remains unclear. Here we describe the clinical presentation, histologic features, and natural history of MLN. We also review the role of supportive treatment and discuss when to deploy immunosuppressive management in MLN.
Collapse
|
19
|
Bomfim GF, Cau SBA, Bruno AS, Fedoce AG, Carneiro FS. Hypertension: a new treatment for an old disease? Targeting the immune system. Br J Pharmacol 2019; 176:2028-2048. [PMID: 29969833 PMCID: PMC6534786 DOI: 10.1111/bph.14436] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/14/2018] [Accepted: 06/24/2018] [Indexed: 12/22/2022] Open
Abstract
Arterial hypertension represents a serious public health problem, being a major cause of morbidity and mortality worldwide. The availability of many antihypertensive therapeutic strategies still fails to adequately treat around 20% of hypertensive patients, who are considered resistant to conventional treatment. In the pathogenesis of hypertension, immune system mechanisms are activated and both the innate and adaptive immune responses play a crucial role. However, what, when and how the immune system is triggered during hypertension development is still largely undefined. In this context, this review highlights scientific advances in the manipulation of the immune system in order to attenuate hypertension and end-organ damage. Here, we discuss the potential use of immunosuppressants and immunomodulators as pharmacological tools to control the activation of the immune system, by non-specific and specific mechanisms, to treat hypertension and improve end-organ damage. Nevertheless, more clinical trials should be performed with these drugs to establish their therapeutic efficacy, safety and risk-benefit ratio in hypertensive conditions. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
| | - Stefany Bruno Assis Cau
- Department of Pharmacology, Institute of Biological ScienceFederal University of Minas GeraisBelo HorizonteMGBrazil
| | - Alexandre Santos Bruno
- Department of Pharmacology, Institute of Biological ScienceFederal University of Minas GeraisBelo HorizonteMGBrazil
| | - Aline Garcia Fedoce
- Department of Pharmacology, Ribeirão Preto Medical SchoolUniversity of São PauloSão PauloBrazil
| | - Fernando S Carneiro
- Department of Pharmacology, Ribeirão Preto Medical SchoolUniversity of São PauloSão PauloBrazil
| |
Collapse
|
20
|
Lehmann AE, Scangas GA, Bergmark RW, El Rassi E, Stankovic KM, Metson R. Periostin and Inflammatory Disease: Implications for Chronic Rhinosinusitis. Otolaryngol Head Neck Surg 2019; 160:965-973. [PMID: 30935271 DOI: 10.1177/0194599819838782] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To provide a comprehensive overview of the emerging role of periostin, an extracellular matrix protein, as a key component in the development, diagnosis, and treatment of patients with chronic rhinosinusitis. DATA SOURCES Medline database. REVIEW METHODS A state of the art review was performed targeting English-language studies investigating the role of periostin in cardiopulmonary, neoplastic, and inflammatory diseases, with emphasis on recent advances in the study of periostin in chronic rhinosinusitis. CONCLUSIONS Periostin has emerged as a novel biomarker and therapeutic target for numerous human pathologies, including cardiac, pulmonary, and neoplastic disease. The upregulation of periostin in chronic rhinosinusitis suggests the potential for similar roles among patients with sinonasal disease. IMPLICATIONS FOR PRACTICE Chronic rhinosinusitis is a widespread disease with major clinical and societal impact. A critical limitation in the current treatment of patients with chronic rhinosinusitis is the absence of clinically relevant biomarkers to guide diagnosis and treatment selection. A review of the literature supports a likely role of periostin as a biomarker of chronic rhinosinusitis, as well as a novel therapeutic target in the future treatment of patients with sinonasal disease.
Collapse
Affiliation(s)
- Ashton E Lehmann
- 1 Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,2 Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - George A Scangas
- 1 Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,2 Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Regan W Bergmark
- 1 Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,2 Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Edward El Rassi
- 1 Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,2 Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Konstantina M Stankovic
- 1 Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,2 Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Ralph Metson
- 1 Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,2 Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Hahn BH, Kono DH. Animal Models in Lupus. DUBOIS' LUPUS ERYTHEMATOSUS AND RELATED SYNDROMES 2019:164-215. [DOI: 10.1016/b978-0-323-47927-1.00014-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
22
|
Khan S, Dar SA, Mandal RK, Jawed A, Wahid M, Panda AK, Lohani M, Mishra BN, Akhter N, Haque S. Angiotensin-Converting Enzyme Gene I/D Polymorphism Is Associated With Systemic Lupus Erythematosus Susceptibility: An Updated Meta-Analysis and Trial Sequential Analysis. Front Physiol 2018; 9:1793. [PMID: 30618805 PMCID: PMC6305102 DOI: 10.3389/fphys.2018.01793] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 11/28/2018] [Indexed: 01/08/2023] Open
Abstract
Angiotensin-converting enzyme (ACE) gene is indispensable for endothelial control and vascular tone regulatory systems, usually affected in Systemic Lupus Erythematosus (SLE). ACE insertion/deletion (I/D) polymorphism may influence the progress of SLE. Earlier studies have investigated this association without any consistency in results. We performed this meta-analysis to evaluate the precise association between ACE I/D polymorphism and SLE susceptibility. The relevant studies were searched until December, 2017 using Medline (PubMed), Google-Scholar and EMBASE search engines. Twenty-five published studies involving 3,308 cases and 4,235 controls were included in this meta-analysis. Statistically significant increased risk was found for allelic (D vs. I: p = 0.007; OR = 1.202, 95% CI = 1.052–1.374), homozygous (DD vs. II: p = 0.025; OR = 1.347, 95% CI = 1.038–1.748), dominant (DD+ID vs. II: p = 0.002; OR = 1.195, 95% CI = 1.070–1.334), and recessive (DD vs. ID+II: p = 0.023; OR = 1.338, 95% CI = 1.042–1.718) genetic models. Subgroup analysis stratified by Asian ethnicity revealed significant risk of SLE in allelic (D vs. I: p = 0.045; OR = 1.238, 95% CI = 1.005–1.525) and marginal risk in dominant (DD+ID vs. II: p = 0.056; OR = 1.192, 95% CI = 0.995–1.428) models; whereas, no association was observed for Caucasian and African population. Publication bias was absent. In conclusion, ACE I/D polymorphism has significant role in overall SLE risk and it can be exploited as a prognostic marker for early SLE predisposition.
Collapse
Affiliation(s)
- Saif Khan
- Department of Basic Science, College of Dental Sciences, University of Ha'il, Ha'il, Saudi Arabia
| | - Sajad A Dar
- The University College of Medical Sciences and GTB, Guru Teg Bahadur Hospital (University of Delhi), New Delhi, India.,Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Raju K Mandal
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Arshad Jawed
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Mohd Wahid
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Aditya K Panda
- Department of Bioscience & Bioinformatics, Khallikote University, Berhampur, India
| | - Mohtashim Lohani
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - B N Mishra
- Department of Biotechnology, Institute of Engineering and Technology, Lucknow, India
| | - Naseem Akhter
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Albaha University, Albaha, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
23
|
Abstract
Most of the attention paid to lupus nephritis, in the medical literature and in clinical trials, has primarily focused on proliferative forms of lupus nephritis (class III and IV lesions), but with lower thresholds to biopsy and rebiopsy patients with lupus, clinicians are encountering more cases with purely mesangial disease (class I and II) or membranous nephropathy patterns (class V). These lesions often will be associated with milder disease courses but still require dedicated follow-up by a nephrologist and focused therapeutic strategies that, at times, will include immunosuppression.
Collapse
Affiliation(s)
- Andrew S Bomback
- Department of Medicine, Division of Nephrology, Columbia University College of Physicians and Surgeons, 622 West 168th Street, PH 4-124, New York, NY 10032, USA.
| |
Collapse
|
24
|
Small HY, Migliarino S, Czesnikiewicz-Guzik M, Guzik TJ. Hypertension: Focus on autoimmunity and oxidative stress. Free Radic Biol Med 2018; 125:104-115. [PMID: 29857140 DOI: 10.1016/j.freeradbiomed.2018.05.085] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 05/22/2018] [Accepted: 05/28/2018] [Indexed: 12/25/2022]
Abstract
Understanding the causal role of the immune and inflammatory responses in hypertension has led to questions regarding the links between hypertension and autoimmunity. Immune pathology in primary hypertension mimics several autoimmune mechanisms observed in the pathogenesis of systemic lupus erythematosus, psoriasis, systemic sclerosis, rheumatoid arthritis and periodontitis. More importantly, the prevalence of hypertension in patients with these autoimmune diseases is significantly increased, when compared to control populations. Clinical and epidemiological evidence is reviewed along with possible mechanisms linking hypertension and autoimmunity. Inflammation and oxidative stress are linked in a self-perpetuating cycle that significantly contributes to the vascular dysfunction and renal damage associated with hypertension. T cell, B cell, macrophage and NK cell infiltration into these organs is essential for this pathology. Effector cytokines such as IFN-γ, TNF-α and IL-17 affect Na+/H+ exchangers in the kidney. In blood vessels, they lead to endothelial dysfunction and loss of nitric oxide bioavailability and cause vasoconstriction. Both renal and vascular effects are, in part, mediated through induction of reactive oxygen species-producing enzymes such as superoxide anion generating NADPH oxidases and dysfunction of anti-oxidant systems. These mechanisms have recently become important therapeutic targets of novel therapies focused on scavenging oxidative (isolevuglandin) modification of neo-antigenic peptides. Effects of classical immune targeted therapies focused on immunosuppression and anti-cytokine treatments are also reviewed.
Collapse
Affiliation(s)
- Heather Y Small
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Serena Migliarino
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Marta Czesnikiewicz-Guzik
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK; Department of Dental Prophylaxis and Experimental Dentistry, Dental School of Jagiellonian University, Krakow, Poland
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK; Department of Internal and Agricultural Medicine, Jagiellonian University Collegium Medicum, Krakow, Poland.
| |
Collapse
|
25
|
Saber S, Mahmoud AAA, Goda R, Helal NS, El-Ahwany E, Abdelghany RH. Perindopril, fosinopril and losartan inhibited the progression of diethylnitrosamine-induced hepatocellular carcinoma in mice via the inactivation of nuclear transcription factor kappa-B. Toxicol Lett 2018; 295:32-40. [PMID: 29859236 DOI: 10.1016/j.toxlet.2018.05.036] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/24/2018] [Accepted: 05/30/2018] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is a major global health problem. Therapeutic interventions of HCC are still limited because of its complicated molecular pathogenesis. Many reports showed that renin-angiotensin system (RAS) contributes to the development of different types of malignancies. Therefore, the present study aimed to examine the effect of RAS inhibition using perindopril (1 mg/kg), fosinopril (2 mg/kg), or losartan (10 mg/kg) on diethylnitrosamine-induced HCC compared to sorafenib (30 mg/kg). The administration of RAS inhibitors resulted in improved liver function and histologic picture with a reduction in AFP levels. These effects found to be mediated through inactivation of NFкB pathway by the inhibition of NFĸB p65 phosphorylation at the Ser536 residue and inhibition of the phosphorylation-induced degradation of NFĸBia. Consequently, expression levels of cyclin D1 mRNA were significantly lowered. In addition, NFкB-induced TNF-α and TGF-β1 levels were reduced leading to lower levels of MMP-2 and VEGF. We concluded that RAS inhibition either through inhibiting the ACE or the blockade of AT1R has the same therapeutic benefit and that the tissue affinity of the ACEIs has no impact on its anti-tumor activity. These results suggest that ACEIs and ARBs can serve as promising candidates for further clinical trials in the management of HCC.
Collapse
Affiliation(s)
- Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa City, Manasoura, Dakahleya, Egypt.
| | - Amr A A Mahmoud
- Department of Pharmacology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt; Department of Pharmacology, Oman Pharmacy Institute, Ministry of Health, Muscat, Oman
| | - Reham Goda
- Department of Microbiology and Immunology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa City, Manasoura, Dakahleya, Egypt
| | - Noha S Helal
- Department of Pathology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Eman El-Ahwany
- Department of Immunology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Rasha H Abdelghany
- Department of Pharmacology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
26
|
Seliga A, Lee MH, Fernandes NC, Zuluaga-Ramirez V, Didukh M, Persidsky Y, Potula R, Gallucci S, Sriram U. Kallikrein-Kinin System Suppresses Type I Interferon Responses: A Novel Pathway of Interferon Regulation. Front Immunol 2018; 9:156. [PMID: 29456540 PMCID: PMC5801412 DOI: 10.3389/fimmu.2018.00156] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/17/2018] [Indexed: 01/13/2023] Open
Abstract
The Kallikrein–Kinin System (KKS), comprised of kallikreins (klks), bradykinins (BKs) angiotensin-converting enzyme (ACE), and many other molecules, regulates a number of physiological processes, including inflammation, coagulation, angiogenesis, and control of blood pressure. In this report, we show that KKS regulates Type I IFN responses, thought to be important in lupus pathogenesis. We used CpG (TLR9 ligand), R848 (TLR7 ligand), or recombinant IFN-α to induce interferon-stimulated genes (ISGs) and proteins, and observed that this response was markedly diminished by BKs, klk1 (tissue kallikrein), or captopril (an ACE inhibitor). BKs significantly decreased the ISGs induced by TLRs in vitro and in vivo (in normal and lupus-prone mice), and in human PBMCs, especially the induction of Irf7 gene (p < 0.05), the master regulator of Type I IFNs. ISGs induced by IFN-α were also suppressed by the KKS. MHC Class I upregulation, a classic response to Type I IFNs, was reduced by BKs in murine dendritic cells (DCs). BKs decreased phosphorylation of STAT2 molecules that mediate IFN signaling. Among the secreted pro-inflammatory cytokines/chemokines analyzed (IL-6, IL12p70, and CXCL10), the strongest suppressive effect was on CXCL10, a highly Type I IFN-dependent cytokine, upon CpG stimulation, both in normal and lupus-prone DCs. klks that break down into BKs, also suppressed CpG-induced ISGs in murine DCs. Captopril, a drug that inhibits ACE and increases BK, suppressed ISGs, both in mouse DCs and human PBMCs. The effects of BK were reversed with indomethacin (compound that inhibits production of PGE2), suggesting that BK suppression of IFN responses may be mediated via prostaglandins. These results highlight a novel regulatory mechanism in which members of the KKS control the Type I IFN response and suggest a role for modulators of IFNs in the pathogenesis of lupus and interferonopathies.
Collapse
Affiliation(s)
- Alecia Seliga
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Michael Hweemoon Lee
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Nicole C Fernandes
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Viviana Zuluaga-Ramirez
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Marta Didukh
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Yuri Persidsky
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Raghava Potula
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Stefania Gallucci
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Uma Sriram
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
27
|
The immunomodulatory effects of the Enalapril in combination with Benznidazole during acute and chronic phases of the experimental infection with Trypanosoma cruzi. Acta Trop 2017; 174:136-145. [PMID: 28720491 DOI: 10.1016/j.actatropica.2017.07.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 07/03/2017] [Accepted: 07/05/2017] [Indexed: 12/20/2022]
Abstract
Trypanosoma cruzi infection triggers a chronic inflammatory process responsible for the alterations in the extracellular matrix and functionality of the heart. The angiotensin converting enzyme (ACE) inhibitors affects T. cruzi in vitro surveillance and modulates in vivo some inflammatory mediators. In this study, we investigated the treatment with an ACE inhibitor (Enalapril) and the Benznidazole (Bz) in a single and combination therapies (CT) in C57BL/6 mice infected with VL-10 strain of the T. cruzi. Animals were treated during 20days with different doses of Bz (100, 80, 60mg/kg), Enalapril (25, 20, 15mg/kg) and their CT (100+25; 80+20; 60+15mg/kg) and euthanized at 30° (acute) and at 120° (chronic) days post infection. The plasma and heart were processed for immunopathological investigations. Our data shown that Bz and Enalapril controlled, in part, the parasite replication and reduced plasma levels of TNF, CCL2 and CCL5 in the acute and in chronic phase of infection. However, the CT doses reduced in around 20% the inflammatory parameters obtained with the Bz therapy. The CT doses of 100+25 and 80+20mg/kg increased the IL-10 levels and reduced the cardiac inflammation while Bz inhibited the collagen neogenesis in the infection. In conclusion, we assume that the CT administrated in the initial stage of infection, presents a minor immunomodulatory effect when the VL-10 strain of T. cruzi is used. In contrast, Bz and Enalapril in monotherapies persist suggesting a potential protection against cardiac damages during experimental T. cruzi infection.
Collapse
|
28
|
Yung S, Yap DYH, Chan TM. Recent advances in the understanding of renal inflammation and fibrosis in lupus nephritis. F1000Res 2017; 6:874. [PMID: 28663794 PMCID: PMC5473406 DOI: 10.12688/f1000research.10445.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/12/2017] [Indexed: 01/08/2023] Open
Abstract
Lupus nephritis is a potentially reversible cause of severe acute kidney injury and is an important cause of end-stage renal failure in Asians and patients of African or Hispanic descent. It is characterized by aberrant exaggerated innate and adaptive immune responses, autoantibody production and their deposition in the kidney parenchyma, triggering complement activation, activation and proliferation of resident renal cells, and expression of pro-inflammatory and chemotactic molecules leading to the influx of inflammatory cells, all of which culminate in destruction of normal nephrons and their replacement by fibrous tissue. Anti-double-stranded DNA (anti-dsDNA) antibody level correlates with disease activity in most patients. There is evidence that apart from mediating pathogenic processes through the formation of immune complexes, pathogenic anti-dsDNA antibodies can bind to resident renal cells and induce downstream pro-apoptotic, pro-inflammatory, or pro-fibrotic processes or a combination of these. Recent data also highlight the critical role of macrophages in acute and chronic kidney injury. Though clinically effective, current treatments for lupus nephritis encompass non-specific immunosuppression and the anti-inflammatory action of high-dose corticosteroids. The clinical and histological impact of novel biologics targeting pro-inflammatory molecules remains to be investigated. Insight into the underlying mechanisms that induce inflammatory and fibrotic processes in the kidney of lupus nephritis could present opportunities for more specific novel treatment options to improve clinical outcomes while minimizing off-target untoward effects. This review discusses recent advances in the understanding of pathogenic mechanisms leading to inflammation and fibrosis of the kidney in lupus nephritis in the context of established standard-of-care and emerging therapies.
Collapse
Affiliation(s)
- Susan Yung
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong
| | - Desmond YH Yap
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong
| | - Tak Mao Chan
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
29
|
Sepehri Z, Masoumi M, Ebrahimi N, Kiani Z, Nasiri AA, Kohan F, Sheikh Fathollahi M, Kazemi Arababadi M, Asadikaram G. Atorvastatin, Losartan and Captopril Lead to Upregulation of TGF-β, and Downregulation of IL-6 in Coronary Artery Disease and Hypertension. PLoS One 2016; 11:e0168312. [PMID: 28033321 PMCID: PMC5199082 DOI: 10.1371/journal.pone.0168312] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 11/30/2016] [Indexed: 12/29/2022] Open
Abstract
Introduction Coronary artery disease (CAD) and hypertension are the main reasons of ischemic heart diseases (IHDs). Cytokines as the small glycoproteins are the main arm of immune system and manipulate all of the cardiovascular diseases. The aim of the current study was to examine the effects of treatment of hypertension and CAD on serum levels of IL-6, IL-8, TGF-β and TNF-α. Material and Methods This interventional study was performed on the patients with hypertension without CAD (group 1), hypertension and CAD (group 2), CAD but not hypertension (group 3) and without hypertension and CAD as controls (group 4). The patients received routine treatment for hypertension and CAD. Serum levels of IL-6, IL-8, TGF-β and TNF-α were analyzed in the groups treated with various drugs, using ELISA technique. Results With regard to the medications, Atorvastatin, Losartan and Captopril were administered more in patients (groups 1, 2 and 3) than the patients without hypertension and CAD. The results revealed that serum levels of TGF-β and IL-6 were significantly increased and decreased, respectively, in the groups 1, 2 and 3 when compared to group 4. Serum levels of TGF-β were also increased in females in comparison to males in the group 4. Discussion According to the results it seems that Atorvastatin, Losartan and Captopril have reduced inflammation in in vivo conditions via downregulation of IL-6 and upregulation of TGF-β.
Collapse
Affiliation(s)
- Zahra Sepehri
- Department of Internal Medicine, Faculty of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Mohammad Masoumi
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
- * E-mail: ,
| | - Nazanin Ebrahimi
- Department of Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Zohre Kiani
- Student Research Committee, Zabol University of Medical Sciences, Zabol, Iran
- Department of Internal Medicine, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Akbar Nasiri
- Department of Anesthesiology, Zabol University of Medical Sciences, Zabol, Iran
| | - Farhad Kohan
- Student Research Committee, Zabol University of Medical Sciences, Zabol, Iran
| | - Mahmood Sheikh Fathollahi
- Immunology of Infectious Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Social Medicine, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Kazemi Arababadi
- Immunology of Infectious Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Laboratory Sciences, Faculty of Paramedicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Gholamreza Asadikaram
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
30
|
Ward F, Bargman JM. Membranous Lupus Nephritis: The Same, But Different. Am J Kidney Dis 2016; 68:954-966. [DOI: 10.1053/j.ajkd.2016.07.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/06/2016] [Indexed: 01/07/2023]
|
31
|
Brook CD, Maxfield AZ, Stankovic K, Metson RB. The Impact of Angiotensin-Modulating Antihypertensives on Time Interval to Revision Surgery for Nasal Polyps. Otolaryngol Head Neck Surg 2016; 155:1046-1052. [PMID: 27554516 DOI: 10.1177/0194599816663924] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/09/2016] [Accepted: 07/21/2016] [Indexed: 11/15/2022]
Abstract
OBJECTIVE/HYPOTHESIS Angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) have been shown to suppress expression of periostin, a matricellular protein that is markedly elevated in nasal polyp tissue. The purpose of this study was to determine whether use of these antihypertensive agents affects the time to revision sinus surgery in patients with polyp regrowth. STUDY DESIGN Case series with chart review. SETTING Academic medical center. SUBJECTS AND METHODS Records were reviewed for 330 patients who underwent ≥2 operations for chronic sinusitis with nasal polyps from April 1987 through August 2015. The time between surgical interventions was compared with patient demographics and clinical characteristics, including use of ACEIs and ARBs. RESULTS Sixty patients were taking ACEIs or ARBs during the study period, of which 32 had concurrent asthma. The mean interval between polyp operations was 61.0 ± 45.2 months (range, 2-228.6 months). Among patients with asthma (n = 197), the mean time to revision surgery was prolonged by >2 years for those taking ACEIs or ARBs (81.0 vs 54.5 months, P = .006). A similar impact on time to revision surgery was not observed for nonasthmatics taking these medications (61.0 vs 65.2 months, P = .655). CONCLUSION Use of ACEIs and ARBs is associated with an increased time to revision sinus surgery among patients with concurrent nasal polyps and asthma. A possible mechanism of this observed effect is suppression of periostin expression through inhibition of the angiotensin pathway.
Collapse
Affiliation(s)
- Christopher D Brook
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA .,Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA
| | - Alice Z Maxfield
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA.,Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA
| | - Konstantina Stankovic
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA.,Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA
| | - Ralph B Metson
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA.,Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
32
|
Tselios K, Gladman DD, Su J, Urowitz MB. Does Renin-Angiotensin System Blockade Protect Lupus Nephritis Patients From Atherosclerotic Cardiovascular Events? A Case-Control Study. Arthritis Care Res (Hoboken) 2016; 68:1497-504. [DOI: 10.1002/acr.22857] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 01/11/2016] [Accepted: 01/26/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Konstantinos Tselios
- Centre for Prognosis Studies in Rheumatic Diseases, Toronto Lupus Clinic, University Health Network; Toronto Ontario Canada
| | - Dafna D. Gladman
- Centre for Prognosis Studies in Rheumatic Diseases, Toronto Lupus Clinic, University Health Network; Toronto Ontario Canada
| | - Jiandong Su
- Centre for Prognosis Studies in Rheumatic Diseases, Toronto Lupus Clinic, University Health Network; Toronto Ontario Canada
| | - Murray B. Urowitz
- Centre for Prognosis Studies in Rheumatic Diseases, Toronto Lupus Clinic, University Health Network; Toronto Ontario Canada
| |
Collapse
|
33
|
Oliveira S, Lopes L, Damazo A, Albuquerque D. Enalapril e captopril revertem o edema e a hiperplasia renais causados pelo antimoniato de N-metilglucamina em camundongos C57BL/6. ARQ BRAS MED VET ZOO 2016. [DOI: 10.1590/1678-4162-8925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
| | - L.L. Lopes
- Universidade Federal de Mato Grosso, Brazil
| | | | | |
Collapse
|
34
|
Almeida LC, Muraro LS, Albuquerque DA. Enhancement of anti-OVA IgG2c production in vivo by enalapril. ACTA ACUST UNITED AC 2016; 49:S0100-879X2016000800603. [PMID: 27409332 PMCID: PMC4954734 DOI: 10.1590/1414-431x20165215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/11/2016] [Indexed: 11/22/2022]
Abstract
Angiotensin-converting enzyme (ACE) inhibitors have non-hemodynamic, pleiotropic effects on the immune response. The effects of ACE inhibitors on the production of cytokines and T-cell functions are well established. However, little is known on the effects of these medicines on humoral response to foreign antigens. In this study, we investigated the effect of enalapril treatment on ovalbumin (OVA)-specific IgG1 and IgG2c production in mice determined by ELISA. Two groups of 8-week-old C57BL/6 females mice (3-4/group) were subcutaneously immunized with OVA (10 μg/animal) in presence of Alhydrogel (1 mg/mouse) and boosted at day 21. The mice were treated with enalapril (5 mg/kg daily, po) or were left without treatment for one month. The animals were bled from the orbital plexus on days 0, 7, 14, 21, and 28 after the first immunization and the sera were stored at -20°C until usage. OVA-specific serum IgG1 and IgG2c were determined by ELISA using serum from each individual animal. The results showed that enalapril significantly increased anti-OVA serum IgG2c in the secondary response without affecting IgG1 synthesis. These data expand our understanding on the properties of enalapril on the immune response, including antibody production.
Collapse
Affiliation(s)
- L C Almeida
- Departamento de Biomedicina, Centro Universitário Cândido Rondon, Cuiabá, MT, Brasil
| | - L S Muraro
- Departamento de Patologia Clínica e Parasitologia, Faculdade de Medicina Veterinária, Universidade de Cuiabá, Cuiabá, MT, Brasil
| | - D A Albuquerque
- Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Cuiabá, MT, Brasil
| |
Collapse
|
35
|
Teplitsky V, Shoenfeld Y, Tanay A. The renin-angiotensin system in lupus: physiology, genes and practice, in animals and humans. Lupus 2016; 15:319-25. [PMID: 16830877 DOI: 10.1191/0961203306lu2306rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Although multiple studies suggest a potential role for angiotensin II in inflammation, most were performed either in vitro or in animals with non-immune-complex-mediated diseases. Extrapolation of these findings to humans, particularly patients with lupus, which involves multiple immunoregulatory pathways, is unclear. In autoimmune-prone MRL/lpr mice, angiotensin-converting-enzyme (ACE) inhibition improved survival although to a lesser degree than cyclophosphamide and diminished the glomerular histopathologic damage, proteinuria, lymphoid hyperplasia, dermatitis, and hypergammaglobulinemia, with a reduction in TGF-beta1 and beta 2 expression in the kidneys and renal chemokine mRNA expression. Spleen levels of IL-4 and IL-10 were also reduced. Uncontrolled studies in patients with treatment-refractory lupus nephritis showed a significant reduction in proteinuria with ACE-inhibitors and Angiotensin receptor blockers treatment. The ‘masking’ effect of ACE-inhibitors should be taken into consideration, as an exacerbation of lupus nephritis may be missed when estimated by the magnitude of proteinuria, which is decreased by these treatments. No single ACE genotype was consistently associated with subsets of SLE patients. In retrospective analyses, ACE-inhibitor use predicted a favourable outcome in 94 cases of pauci-immune vasculitis. The attenuating effect of angiotensin II inhibitors on the progression of chronic renal disease is well recognized. The data on the role of this intervention in lupus is limited.
Collapse
Affiliation(s)
- V Teplitsky
- Immunology/Allergy Unit, Rabin Medical Center, Petah Tiqwa, Israel.
| | | | | |
Collapse
|
36
|
Wu JY, Lee MTG, Lee SH, Lee SH, Tsai YW, Hsu SC, Chang SS, Lee CC. Angiotensin-Converting Enzyme Inhibitors and Active Tuberculosis: A Population-Based Study. Medicine (Baltimore) 2016; 95:e3579. [PMID: 27175655 PMCID: PMC4902497 DOI: 10.1097/md.0000000000003579] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Numerous epidemiological data suggest that the use of angiotensin-converting enzyme inhibitors (ACEis) can improve the clinical outcomes of pneumonia. Tuberculosis (TB) is an airborne bacteria like pneumonia, and we aimed to find out whether the use of ACEis can decrease the risk of active TB.We conducted a nested case-control analysis by using a 1 million longitudinally followed cohort, from Taiwan national health insurance research database. The rate ratios (RRs) for TB were estimated by conditional logistic regression, and adjusted using a TB-specific disease risk score (DRS) with 71 TB-related covariates.From January, 1997 to December, 2011, a total of 75,536 users of ACEis, and 7720 cases of new active TB were identified. Current use (DRS adjusted RR, 0.87 [95% CI, 0.78-0.97]), but not recent and past use of ACEis, was associated with a decrease in risk of active TB. Interestingly, it was found that chronic use (>90 days) of ACEis was associated with a further decrease in the risk of TB (aRR, 0.74, [95% CI, 0.66-0.83]). There was also a duration response effect, correlating decrease in TB risk with longer duration of ACEis use. The decrease in TB risk was also consistent across all patient subgroups (age, sex, heart failure, cerebrovascular diseases, myocardial infraction, renal diseases, and diabetes) and patients receiving other cardiovascular medicine.In this large population-based study, we found that subjects with recent and chronic use of ACEis were associated with decrease in TB risk.
Collapse
Affiliation(s)
- Jiunn-Yih Wu
- From the Department of Emergency Medicine, Chang Gung Memorial Hospital, Keelung; Chang Gung University College of Medicine, Taoyuan (J-YW, S-CH); Department of Emergency Medicine, National Taiwan University Hospital (M-TGL, S-HL, C-CL); Department of Rehabilitation and Physical Medicine, Taipei Veteran General Hospital (S-HL); Department of Medicine, College of Medicine, National Yang Ming University, Taipei (S-HL); Department of Family Medicine, Chang Gung Memorial Hospital; Chang Gung University College of Medicine (Y-WT, S-SC); Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan (S-SC); and Department of Emergency Medicine and Department of General Medicine, National Taiwan University Hospital Yun-lin Branch, Douliou (C-CL), Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Taylor EB, Ryan MJ. Understanding mechanisms of hypertension in systemic lupus erythematosus. Ther Adv Cardiovasc Dis 2016; 11:1753944716637807. [PMID: 26985016 PMCID: PMC5065379 DOI: 10.1177/1753944716637807] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder that predominately affects women of reproductive age. Hypertension is an important cardiovascular risk factor that is prevalent in this patient population. Despite the high incidence of hypertension in women with SLE, the pathophysiological mechanisms underlying the development of hypertension remain poorly understood. This review will focus on disease-related factors, including inflammation, autoantibodies, and sex hormones that may contribute to hypertension in patients with SLE. In addition, we will highlight studies performed by our laboratory using the female NZBWF1 (F1 hybrid of New Zealand Black and New Zealand White strains) mouse model, a spontaneous model of SLE that mimics human disease and develops hypertension and renal injury. Specifically, using female NZBWF1 mice, we have demonstrated that multiple factors contribute to the pathogenesis of hypertension, including the inflammatory cytokine, tumor necrosis factor (TNF)-α, oxidative stress, as well as B-cell hyperactivity and autoantibody production.
Collapse
Affiliation(s)
- Erin B Taylor
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| |
Collapse
|
38
|
Association between Angiotensin I-Converting Enzyme Insertion/Deletion Polymorphism and Prognosis of Kidney Transplantation: A Meta-Analysis. PLoS One 2015; 10:e0127320. [PMID: 26000752 PMCID: PMC4441456 DOI: 10.1371/journal.pone.0127320] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 04/13/2015] [Indexed: 01/06/2023] Open
Abstract
PURPOSE Angiotensin I-converting enzyme (ACE) is crucial in the renin-angiotensin-aldosterone system. ACE insertion/deletion (I/D) polymorphism is a common genetic variation of this gene and is associated with several disease phenotypes. However, the results of published studies on the influence of this polymorphism on renal transplantation are inconsistent. Therefore, a meta-analysis was performed to evaluate the association between ACE I/D polymorphism and prognosis of kidney transplantation. METHODS A meta-analysis was performed based on 21 case-control studies from 12 publications (1497 cases and 2029 controls) and 10 studies with quantitative values from 5 publications (814 patients). Pooled odds ratios (ORs) and weighted mean differences (WMDs) with their corresponding 95% confidence intervals (CIs) were used to estimate associations. RESULTS ACE I/D polymorphism was found to be associated with acute rejection (AR) in genotypes DD+ID versus II (OR = 1.62, 95% CI = 1.14-2.29) and with serum creatinine concentration after renal transplantation in genotypes DD versus ID (WMD = 13.12, 95% CI = 8.09-18.16). Stratified analysis revealed that recipients transplanted within a year had higher serum creatinine concentrations in the DD versus ID model. No significant association was found between hypertension and ACE I/D polymorphism. CONCLUSION ACE I/D polymorphism is associated with AR and allograft function after kidney transplantation.
Collapse
|
39
|
Park S, Park M, Kim BH, Lee JE, Park HJ, Lee SH, Park CG, Kim MH, Kim R, Kim EH, Heo CY, Choy YB. Acute suppression of TGF-ß with local, sustained release of tranilast against the formation of fibrous capsules around silicone implants. J Control Release 2014; 200:125-37. [PMID: 25528612 DOI: 10.1016/j.jconrel.2014.12.021] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 11/16/2014] [Accepted: 12/16/2014] [Indexed: 12/19/2022]
Abstract
We propose the acute, local suppression of transforming growth factor beta (TGF-ß), a major profibrotic cytokine, to reduce fibrosis around silicone implants. To this end, we prepared silicone implants that were able to release tranilast, a TGF-ß inhibitor, in a sustained manner for 5 days or 15 days. We performed histologic and immunohistochemical analyses for 12 weeks after the implantation of the implants in living rats. The capsule thicknesses and collagen densities significantly decreased compared with those around the non-treated silicone implants. Notably, early suppression of TGF-ß affected the fibrogenesis that actually occurs at the late stage of wound healing. This change may be ascribed to the decrease in monocyte recruitment mediated by early TGF-ß during the acute inflammatory reaction. Thus, a significant decrease in differentiated macrophages was observed along with a decrease in the quantity of TGF-ß and fibroblasts during the subsequent inflammation stage; these changes led to a diminished fibrotic capsule formation.
Collapse
Affiliation(s)
- Subin Park
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Min Park
- Interdisciplinary Program in Bioengineering, College of Engineering, Seoul National University, Seoul 152-742, Republic of Korea
| | - Byung Hwi Kim
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Ji Eun Lee
- Interdisciplinary Program in Bioengineering, College of Engineering, Seoul National University, Seoul 152-742, Republic of Korea
| | - Hyo Jin Park
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam 463-707, Republic of Korea
| | - Seung Ho Lee
- Interdisciplinary Program in Bioengineering, College of Engineering, Seoul National University, Seoul 152-742, Republic of Korea
| | - Chun Gwon Park
- Institute of Medical & Biological Engineering, Medical Research Center, Seoul National University, Seoul 110-799, Republic of Korea
| | - Myung Hun Kim
- Interdisciplinary Program in Bioengineering, College of Engineering, Seoul National University, Seoul 152-742, Republic of Korea
| | - Ryul Kim
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Eun Hee Kim
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam 463-707, Republic of Korea
| | - Chan Yeong Heo
- Department of Plastic and Reconstructive Surgery, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam 463-707, Republic of Korea.
| | - Young Bin Choy
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; Interdisciplinary Program in Bioengineering, College of Engineering, Seoul National University, Seoul 152-742, Republic of Korea; Institute of Medical & Biological Engineering, Medical Research Center, Seoul National University, Seoul 110-799, Republic of Korea.
| |
Collapse
|
40
|
Tselios K, Koumaras C, Urowitz MB, Gladman DD. Do current arterial hypertension treatment guidelines apply to systemic lupus erythematosus patients? A critical appraisal. Semin Arthritis Rheum 2014; 43:521-5. [DOI: 10.1016/j.semarthrit.2013.07.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 06/12/2013] [Accepted: 07/08/2013] [Indexed: 12/31/2022]
|
41
|
Ackerman Z, Grozovski M, Oron-Herman M, Rosenthal T, Sela BA, Amir G. Effects of Antihypertensive and Triglyceride Lowering Agents on Splenocyte Apoptosis in Rats with Fatty Liver. Basic Clin Pharmacol Toxicol 2013; 113:37-42. [DOI: 10.1111/bcpt.12067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Accepted: 02/25/2013] [Indexed: 01/20/2023]
Affiliation(s)
- Zvi Ackerman
- Department of Medicine; Hadassah-Hebrew University Medical Center; Jerusalem; Israel
| | | | | | - Talma Rosenthal
- Sackler Faculty of Medicine; Tel Aviv University; Tel Aviv; Israel
| | - Ben-Ami Sela
- Institute of Chemical Pathology; Sheba Medical Center; Tel Hashomer; Israel
| | - Gail Amir
- Department of Pathology; Hadassah-Hebrew University Medical Center; Jerusalem; Israel
| |
Collapse
|
42
|
Singh RR, Yang JQ, Kim PJ, Halder RC. Germline deletion of β2 microglobulin or CD1d reduces anti-phospholipid antibody, but increases autoantibodies against non-phospholipid antigens in the NZB/W F1 model of lupus. Arthritis Res Ther 2013; 15:R47. [PMID: 23531237 PMCID: PMC3672782 DOI: 10.1186/ar4206] [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: 11/16/2012] [Accepted: 03/19/2013] [Indexed: 11/25/2022] Open
Abstract
Introduction β2-microglobulin (β2m) is required for the surface expression of MHC class I and class I-like proteins such as CD1d, Qa1 and neonatal Fc receptor (FcRn), all of which may impact the development of autoimmunity. Since CD1d is known to bind and present phospholipid antigens to T cells, we asked if the deficiency of β2m or CD1d will impact the development of anti-phospholipid antibodies as compared to other aspects of lupus autoimmunity. Methods We introgressed the β2m-null genotype onto the NZB and NZW backgrounds for 12 to 14 generations to generate genetically lupus-susceptible (NZB/NZW)F1 (BWF1) mice that are β2m-deficient (β2m°). Circulating immunoglobulins (Ig), rheumatoid factor (RF), anti-DNA and anti-cardiolipin (anti-CL) antibodies, and renal disease were analyzed in these and CD1d-deficient (CD1d°) BWF1 mice that we had previously generated. Results Whereas β2m° BWF1 mice had reduced serum IgG, they had increased mortality, nephritis, serum IgG anti-DNA antibody and RF as compared to heterozygous and wild-type littermates. These effects were recapitulated in CD1d° BWF1 mice, except that they also had increased serum IgG as compared to control littermates. Intriguingly, both β2m° and CD1d° mice had lower serum anti-CL antibody levels than in control littermates. Such CD1d dependence of anti-CL antibody production is not mediated by CD1d/glycolipid-reactive iNKT cells, as these cells reduced the production of RF and anti-DNA antibodies but had no effect on anti-CL antibodies. Conclusions We report a novel dichotomous role of β2m and CD1d, whereby these molecules differently regulate autoimmunity against phospholipid versus non-phospholipid autoantigens.
Collapse
|
43
|
Yen TH, Yang HY, Yeh YH, Chu PH, Wen CJ, Fu JF, Wang IK, Liang CC, Chang CT, Chen KH, Tian YC, Hung CC, Lin JL, Yang CW. Aliskiren attenuates proteinuria in mice with lupus nephritis by a blood pressure-independent mechanism. Lupus 2012; 22:180-9. [PMID: 23257405 DOI: 10.1177/0961203312471871] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This study revealed that low-dose aliskiren treatment could attenuate proteinuria by interrupting the renin-angiotensin system in mice with lupus nephritis, and the beneficial effect was beyond blood pressure control. An in and ex vivo fluorescence imaging (using a non-invasion in vivo imaging system) showed intense labeling of renin in the kidneys of female MRL/lpr mice. In the study, Alzet mini-osmotic pumps were implanted into 6-week-old female MRL/lpr mice. Pumps were filled with either phosphate-buffered saline or a solution of aliskiren dissolved in phosphate-buffered saline (20 mg/kg/day) and replaced at 28-day intervals. Mice were sacrificed at four and eight weeks. To label cells for DNA synthesis, bromodeoxyuridine (BrdU) (50 mg/kg) was injected intraperitoneally an hour prior to sacrifice. The level of renin inhibition was adequate, as aliskiren-treated mice demonstrated higher renal renin mRNA expression than controls (p < 0.05). Although there were no significant differences in the systolic blood pressure (control versus aliskiren-treated: 127.20 ± 4.44 mmHg versus 103.80 ± 7.40 mmHg, p > 0.05) and heart rate (control versus aliskiren-treated: 680.50 ± 11.71 versus 647.80 ± 13.90, p > 0.05) of both groups after eight weeks, there was significant reduction of inflammatory cytokines (transforming growth factor-beta1, regulated on activation normal T cell expressed, monocyte chemoattractant protein-1 and osteopontin, p < 0.05), reduction of innate immunity (toll-like receptor 7, p < 0.05), as well as a reduction of glomerular proliferation and inflammation (BrdU-, CD45-, CD3- and F4/80-positive glomerular cells, p < 0.01) after aliskiren infusion, which might translate into an improvement in proteinuria (control versus aliskiren-treated: 493.7 versus 843.7 mg/g, p < 0.01) or weight gain (control versus aliskiren-treated: 5.65 ± 1.61 versus 8.67 ± 0.97%, p < 0.05).
Collapse
Affiliation(s)
- T-H Yen
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, 199 Tung Hwa North Rd., Taipei, Taiwan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
During the course of systemic lupus erythematosus (SLE) 30-90% of patients develop a renal manifestation which has proven to be decisive for morbidity and mortality. Histologically six different classes have been described leading to different treatment strategies. In mesangial proliferative lupus nephritis (class II) extrarenal manifestations determine the immunosuppressive treatment. However, in class III and IV (focal or diffuse proliferative manifestation) cyclophosphamide or possibly mycophenolate mofetil (MMF) as an alternative is necessary. In membranous lupus nephritis (class V) dual renin-angiotensin aldosterone (RAAS) blockade is most important. With class I (minimal mesangial lupus nephritis) and class VI (sclerosis) no immunosuppressive therapy is needed. New treatment options concentrate on B-cell depletion, inhibition of cytokines and co-stimulatory molecules. Recently, for the first time in SLE, a monoclonal antibody (belimumab) against B lymphocyte-stimulating factor (Blys) has been approved for treatment in combination with standard therapy.
Collapse
|
45
|
Recent advances in the treatment of lupus nephritis. Clin Exp Nephrol 2011; 16:202-13. [PMID: 22057583 DOI: 10.1007/s10157-011-0556-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2011] [Accepted: 10/17/2011] [Indexed: 10/15/2022]
Abstract
Lupus nephritis is a common complication of systemic lupus erythematosus (SLE) which is associated with significant morbidity and mortality. The concept of two phases of therapy for lupus nephritis, such as an induction phase and a maintenance phase, is widely accepted. Since the renal involvement in SLE is heterogeneous, the treatment of lupus nephritis is governed by its pathological type and ranges from nonspecific measures, such as maintenance of adequate blood pressure control and blockade of the renin-angiotensin-aldosterone system, to the use of immunosuppressive agents. Cyclophosphamide (CYC) in combination with prednisone has been the standard method of treatment of the proliferative forms of lupus nephritis. However, the high rates of progression to end-stage renal disease coupled with the adverse effects of CYC and prednisone have led to an intensive search for more effective and less toxic therapies for lupus nephritis. We review the options available for the treatment of proliferative and membranous lupus nephritis and summarize the results of recently published clinical trials that add new perspectives to the management of kidney disease in SLE.
Collapse
|
46
|
Lundberg S, Lundahl J, Gunnarsson I, Sundelin B, Jacobson SH. Soluble interleukin-2 receptor alfa predicts renal outcome in IgA nephropathy. Nephrol Dial Transplant 2011; 27:1916-23. [PMID: 21940483 DOI: 10.1093/ndt/gfr554] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Both systemic and mucosal IgA production are controlled by T lymphocytes and infiltrating T lymphocytes are involved in the progression of interstitial fibrosis in chronic kidney disease (CKD). Since the concentration of soluble interleukin-2 receptor alfa (sIL-2Ra) reflects the degree of T cell activation over time, we studied the impact of interleukin-2 receptor alfa levels on disease progression in patients with biopsy-proven IgA nephropathy (IgAN), a disease in which 20-30% of the patients progress to end-stage renal failure. METHODS sIL-2Ra plasma levels were measured in 194 patients (median age 39 years, 70% men) and 84 matched controls. One hundred and seventy-nine of the patients, with an estimated glomerular filtration rate (GFR) of ≥15 mL/min/1.73m(2) at baseline (CKD Stages 1-4), were followed for up to 15 years (median 52 months; range 12-188). sIL-2Ra was evaluated as a risk marker for severe renal progression, here defined by the development of CKD Stage 5 (GFR <15 mL/min/1.73m(2)), a 50% decline in GFR during the follow-up period or a 30% GFR decline within 5 years of follow-up. In 51 patients, upon whom a renal biopsy had been performed within 2 years of IL2-Ra measurement, the biopsies were scored according to the Oxford classification. The correlations between the histopathological findings and the sIL-2Ra levels were examined. RESULTS sIL2-Ra levels were significantly higher in patients than in controls (P < 0.001). sIL-2Ra levels in the upper third tertile predicted a severe renal outcome, even after adjustment for the main clinical risk factors: time average albuminuria and GFR at baseline (Relative risk 5.35, P < 0.001). sIL-2Ra levels also correlated significantly to the yearly GFR slope (β = -0.24, P = 0.01). According to the Oxford classification, the presence of >25% tubular atrophy/interstitial fibrosis (T1-2) was associated with higher sIL-2Ra levels, after adjustment for serum creatinine levels, if analysed within 4 months [n = 24, odds ratio (OR) 1.0, P = 0.044] or within 2 years from the kidney biopsy (n = 51, OR 1.0, P = 0.017). CONCLUSIONS The plasma levels of sIL-2Ra were predictive of long-term renal disease progression in a large cohort of patients with biopsy-proven IgAN. Further studies are warranted to evaluate if sIL-2Ra levels can feasibly contribute in the monitoring of effects of treatment, aimed to prevent the progression of interstitial fibrosis and progressive glomerulosclerosis in IgAN.
Collapse
Affiliation(s)
- Sigrid Lundberg
- Nephrology Unit, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
47
|
Venegas-Pont M, Mathis KW, Iliescu R, Ray WH, Glover PH, Ryan MJ. Blood pressure and renal hemodynamic responses to acute angiotensin II infusion are enhanced in a female mouse model of systemic lupus erythematosus. Am J Physiol Regul Integr Comp Physiol 2011; 301:R1286-92. [PMID: 21900645 DOI: 10.1152/ajpregu.00079.2011] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Inflammation and immune system dysfunction contributes to the development of cardiovascular and renal disease. Systemic lupus erythematosus (SLE) is a chronic autoimmune inflammatory disorder that carries a high risk for both renal and cardiovascular disease. While hemodynamic changes that may contribute to increased cardiovascular risk have been reported in humans and animal models of SLE, renal hemodynamics have not been widely studied. The renin-angiotensin system (RAS) plays a central role in renal hemodynamic control, and although RAS blockade is a common therapeutic strategy, the role of RAS in hemodynamic function during SLE is not clear. This study tested whether mean arterial pressure (MAP) and renal hemodynamic responses to acute infusions of ANG II in anesthetized animals were enhanced in an established female mouse model of SLE (NZBWF1). Baseline MAP was not different between anesthetized SLE and control (NZWLacJ) mice, while renal blood flow (RBF) was significantly lower in mice with SLE. SLE mice exhibited an enhanced pressor response and greater reduction in RBF after ANG II infusion. An acute infusion of the ANG II receptor blocker losartan increased RBF in control mice but not in mice with SLE. Renin and ANG II type 1 receptor expression was significantly lower, and ANG II type 2 receptor expression was increased in the renal cortex from SLE mice compared with controls. These data suggest that there are fewer ANG II receptors in the kidneys from mice with SLE but that the existing receptors exhibit an enhanced sensitivity to ANG II.
Collapse
Affiliation(s)
- Marcia Venegas-Pont
- Department of Physiology and Biophysics and Center for Excellence in Cardiovascular Renal Research, University of Mississippi Medical Center, Jackson, Mississippi 39216-4505, USA
| | | | | | | | | | | |
Collapse
|
48
|
The role of EMT in renal fibrosis. Cell Tissue Res 2011; 347:103-16. [PMID: 21845400 DOI: 10.1007/s00441-011-1227-1] [Citation(s) in RCA: 245] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 07/22/2011] [Indexed: 02/03/2023]
Abstract
It is clear that the well-described phenomenon of epithelial-mesenchymal transition (EMT) plays a pivotal role in embryonic development, wound healing, tissue regeneration, organ fibrosis and cancer progression. EMTs have been classified into three subtypes based on the functional consequences and biomarker context in which they are encountered. This review will highlight findings on type II EMT as a direct contributor to the kidney myofibroblast population in the development of renal fibrosis, specifically in diabetic nephropathy, the signalling molecules and the pathways involved in type II EMT and changes in the expression of specific miRNA with the EMT process. These findings have provided new insights into the activation and development of EMT during disease processes and may lead to possible therapeutic interventions to suppress EMTs and potentially reverse organ fibrosis.
Collapse
|
49
|
Hills CE, Squires PE. The role of TGF-β and epithelial-to mesenchymal transition in diabetic nephropathy. Cytokine Growth Factor Rev 2011; 22:131-9. [PMID: 21757394 DOI: 10.1016/j.cytogfr.2011.06.002] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Transforming Growth Factor-beta (TGF-β) is a pro-sclerotic cytokine widely associated with the development of fibrosis in diabetic nephropathy. Central to the underlying pathology of tubulointerstitial fibrosis is epithelial-to-mesenchymal transition (EMT), or the trans-differentiation of tubular epithelial cells into myofibroblasts. This process is accompanied by a number of key morphological and phenotypic changes culminating in detachment of cells from the tubular basement membrane and migration into the interstitium. Ultimately these cells reside as activated myofibroblasts and further exacerbate the state of fibrosis. A large body of evidence supports a role for TGF-β and downstream Smad signalling in the development and progression of renal fibrosis. Here we discuss a role for TGF-β as the principle effector in the development of renal fibrosis in diabetic nephropathy, focusing on the role of the TGF-β1 isoform and its downstream signalling intermediates, the Smad proteins. Specifically we review evidence for TGF-β1 induced EMT in both the proximal and distal regions of the nephron and describe potential therapeutic strategies that may target TGF-β1 activity.
Collapse
Affiliation(s)
- Claire E Hills
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK.
| | | |
Collapse
|
50
|
Abstract
IGF-1 (insulin-like growth factor-1) plays a unique role in the cell protection of multiple systems, where its fine-tuned signal transduction helps to preserve tissues from hypoxia, ischaemia and oxidative stress, thus mediating functional homoeostatic adjustments. In contrast, its deprivation results in apoptosis and dysfunction. Many prospective epidemiological surveys have associated low IGF-1 levels with late mortality, MI (myocardial infarction), HF (heart failure) and diabetes. Interventional studies suggest that IGF-1 has anti-atherogenic actions, owing to its multifaceted impact on cardiovascular risk factors and diseases. The metabolic ability of IGF-1 in coupling vasodilation with improved function plays a key role in these actions. The endothelial-protective, anti-platelet and anti-thrombotic activities of IGF-1 exert critical effects in preventing both vascular damage and mechanisms that lead to unstable coronary plaques and syndromes. The pro-survival and anti-inflammatory short-term properties of IGF-1 appear to reduce infarct size and improve LV (left ventricular) remodelling after MI. An immune-modulatory ability, which is able to suppress 'friendly fire' and autoreactivity, is a proposed important additional mechanism explaining the anti-thrombotic and anti-remodelling activities of IGF-1. The concern of cancer risk raised by long-term therapy with IGF-1, however, deserves further study. In the present review, we discuss the large body of published evidence and review data on rhIGF-1 (recombinant human IGF-1) administration in cardiovascular disease and diabetes, with a focus on dosage and safety issues. Perhaps the time has come for the regenerative properties of IGF-1 to be assessed as a new pharmacological tool in cardiovascular medicine.
Collapse
|