1
|
Fan D, Wu R. Mechanisms of the septic heart: From inflammatory response to myocardial edema. J Mol Cell Cardiol 2024; 195:73-82. [PMID: 39142438 DOI: 10.1016/j.yjmcc.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 08/10/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Sepsis-induced myocardial dysfunction (SIMD), also known as sepsis-induced cardiomyopathy (SICM), is linked to significantly increased mortality. Despite its clinical importance, effective therapies for SIMD remain elusive, largely due to an incomplete understanding of its pathogenesis. Over the past five decades, research involving both animal models and human studies has highlighted several pathogenic mechanisms of SICM, yet many aspects remain unexplored. Initially thought to be primarily driven by inflammatory cytokines, current research indicates that these alone are insufficient for the development of cardiac dysfunction. Recent studies have brought attention to additional mechanisms, including excessive nitric oxide production, mitochondrial dysfunction, and disturbances in calcium homeostasis, as contributing factors in SICM. Emerging clinical evidence has highlighted the significant role of myocardial edema in the pathogenesis of SICM, particularly its association with cardiac remodeling in septic shock patients. This review synthesizes our current understanding of SIMD/SICM, focusing on myocardial edema's contribution to cardiac dysfunction and the critical role of the bradykinin receptor B1 (B1R) in altering myocardial microvascular permeability, a potential key player in myocardial edema development during sepsis. Additionally, this review briefly summarizes existing therapeutic strategies and their challenges and explores future research directions. It emphasizes the need for a deeper understanding of SICM to develop more effective treatments.
Collapse
Affiliation(s)
- Dihan Fan
- Psychiatric Genetics Group, McGill University, Canada
| | - Rongxue Wu
- Department of Medicine, Section of Cariology, Biological Sciences Division, The University of Chicago, IL, United States.
| |
Collapse
|
2
|
Siti-Zubaidah MZ, Harafinova HS, Liba AN, Nordin ML, Hambali KA, Siti HN. Exploring bradykinin: A common mediator in the pathophysiology of sepsis and atherosclerotic cardiovascular disease. Vascul Pharmacol 2024; 156:107414. [PMID: 39089528 DOI: 10.1016/j.vph.2024.107414] [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: 04/13/2024] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Sepsis and atherosclerotic cardiovascular disease (ASCVD) are major health challenges involving complex processes like inflammation, renin-angiotensin system (RAS) dysregulation, and thrombosis. Despite distinct clinical symptoms, both conditions share mechanisms mediated by bradykinin. This review explores bradykinin's role in inflammation, RAS modulation, and thrombosis in sepsis and ASCVD. In sepsis, variable kininogen-bradykinin levels may correlate with disease severity and progression, though the effect of bradykinin receptor modulation on inflammation remains uncertain. RAS activation is present in both diseases, with sepsis showing variable or low levels of Ang II, ACE, and ACE2, while ASCVD consistently exhibits elevated levels. Bradykinin may act as a mediator for ACE2 and AT2 receptor effects in RAS regulation. It may influence clotting and fibrinolysis in sepsis-associated coagulopathy, but evidence for an antithrombotic effect in ASCVD is insufficient. Understanding bradykinin's role in these shared pathologies could guide therapeutic and monitoring strategies and inform future research.
Collapse
Affiliation(s)
- Mohd Zahari Siti-Zubaidah
- Department of Anaesthesia and Intensive Care, National Heart Institute, Jalan Tun Razak, 50400 Kuala Lumpur, Malaysia.
| | - Harman-Shah Harafinova
- Department of Internal Medicine, Faculty of Medicine, Universiti Sultan Zainal Abidin, Jalan Sultan Mahmud, 20400 Kuala Terengganu, Terengganu, Malaysia.
| | - Abdullahi Nuradeen Liba
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Malaysia Kelantan, Pengkalan Chepa, Kota Bharu, 16100, Kelantan, Malaysia
| | - Muhammad Luqman Nordin
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Malaysia Kelantan, Pengkalan Chepa, Kota Bharu, 16100, Kelantan, Malaysia; Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Kamarul Ariffin Hambali
- Faculty of Earth Science, Universiti Malaysia Kelantan, Jeli, 17600, Kelantan, Malaysia; Animal and Wildlife Research Group, Faculty of Earth Science, Jeli Campus, Universiti Malaysia Kelantan, 17600, Kelantan, Malaysia.
| | - Hawa Nordin Siti
- Department of Pharmacology, Faculty of Medicine, Universiti Sultan Zainal Abidin, Jalan Sultan Mahmud, 20400 Kuala Terengganu, Terengganu, Malaysia.
| |
Collapse
|
3
|
Mun SJ, Cho E, Kim HK, Gil WJ, Yang CS. Enhancing acute inflammatory and sepsis treatment: superiority of membrane receptor blockade. Front Immunol 2024; 15:1424768. [PMID: 39081318 PMCID: PMC11286478 DOI: 10.3389/fimmu.2024.1424768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024] Open
Abstract
Conditions such as acute pancreatitis, ulcerative colitis, delayed graft function and infections caused by a variety of microorganisms, including gram-positive and gram-negative organisms, increase the risk of sepsis and therefore mortality. Immune dysfunction is a characterization of sepsis, so timely and effective treatment strategies are needed. The conventional approaches, such as antibiotic-based treatments, face challenges such as antibiotic resistance, and cytokine-based treatments have shown limited efficacy. To address these limitations, a novel approach focusing on membrane receptors, the initiators of the inflammatory cascade, is proposed. Membrane receptors such as Toll-like receptors, interleukin-1 receptor, endothelial protein C receptor, μ-opioid receptor, triggering receptor expressed on myeloid cells 1, and G-protein coupled receptors play pivotal roles in the inflammatory response, offering opportunities for rapid regulation. Various membrane receptor blockade strategies have demonstrated efficacy in both preclinical and clinical studies. These membrane receptor blockades act as early stage inflammation modulators, providing faster responses compared to conventional therapies. Importantly, these blockers exhibit immunomodulatory capabilities without inducing complete immunosuppression. Finally, this review underscores the critical need for early intervention in acute inflammatory and infectious diseases, particularly those posing a risk of progressing to sepsis. And, exploring membrane receptor blockade as an adjunctive treatment for acute inflammatory and infectious diseases presents a promising avenue. These novel approaches, when combined with antibiotics, have the potential to enhance patient outcomes, particularly in conditions prone to sepsis, while minimizing risks associated with antibiotic resistance and immune suppression.
Collapse
Affiliation(s)
- Seok-Jun Mun
- Department of Bionano Engineering, Hanyang University, Seoul, Republic of Korea
- Center for Bionano Intelligence Education and Research, Hanyang University, Ansan, Republic of Korea
| | - Euni Cho
- Department of Bionano Engineering, Hanyang University, Seoul, Republic of Korea
- Center for Bionano Intelligence Education and Research, Hanyang University, Ansan, Republic of Korea
| | - Hyo Keun Kim
- Center for Bionano Intelligence Education and Research, Hanyang University, Ansan, Republic of Korea
- Department of Molecular and Life Science, Hanyang University, Ansan, Republic of Korea
| | - Woo Jin Gil
- Center for Bionano Intelligence Education and Research, Hanyang University, Ansan, Republic of Korea
- Department of Molecular and Life Science, Hanyang University, Ansan, Republic of Korea
| | - Chul-Su Yang
- Center for Bionano Intelligence Education and Research, Hanyang University, Ansan, Republic of Korea
- Department of Molecular and Life Science, Hanyang University, Ansan, Republic of Korea
- Department of Medicinal and Life Science, Hanyang University, Ansan, Republic of Korea
| |
Collapse
|
4
|
Fernandes Gregnani M, Budu A, Batista RO, Ornellas FH, Estrela GR, Arruda AC, Freitas Lima LC, Kremer JL, Favaroni Mendes LA, Casarini DE, Lotfi CFP, Oyama LM, Bader M, Araújo RC. Kinin B1 receptor modulates glucose homeostasis and physical exercise capacity by altering adrenal catecholamine synthesis and secretion. Mol Cell Endocrinol 2024; 579:112085. [PMID: 37827227 DOI: 10.1016/j.mce.2023.112085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/14/2023]
Abstract
Our group has shown in several papers that kinin B1 receptor (B1R) is involved in metabolic adaptations, mediating glucose homeostasis and interfering in leptin and insulin signaling. Since catecholamines are involved with metabolism management, we sought to evaluate B1R role in catecholamine synthesis/secretion. Using B1R global knockout mice, we observed increased basal epinephrine content, accompanied by decreased hepatic glycogen content and increased glucosuria. When these mice were challenged with maximal intensity exercise, they showed decreased epinephrine and norepinephrine response, accompanied by disturbed glycemic responses to effort and poor performance. This phenotype was related to alterations in adrenal catecholamine synthesis: increased basal epinephrine concentration and reduced norepinephrine content in response to exercise, as well decreased gene expression and protein content of tyrosine hydroxylase and decreased gene expression of dopamine beta hydroxylase and kinin B2 receptor. We conclude that the global absence of B1R impairs catecholamine synthesis, interfering with glucose metabolism at rest and during maximal exercise.
Collapse
Affiliation(s)
- Marcos Fernandes Gregnani
- Department of Byophisics, Federal University of São Paulo, Brazil; Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany.
| | - Alexandre Budu
- Department of Byophisics, Federal University of São Paulo, Brazil
| | | | | | - Gabriel Rufino Estrela
- Department of Medicine, Discipline of Nephrology, Federal University of Sao Paulo, São Paulo, Brazil; Department of Clinical and Experimental Oncology, Discipline of Hematology and Hematotherapy, Federal University of São Paulo, 04037002, São Paulo, Brazil
| | | | | | - Jean Lucas Kremer
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Dulce Elena Casarini
- Department of Medicine, Discipline of Nephrology, Federal University of Sao Paulo, São Paulo, Brazil
| | | | | | - Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10117, Berlin, Germany; Max Delbrück Center of Molecular Medicine, Charité University Medicine, Charitéplatz 1, 10117, Berlin, Germany; Institute for Biology, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | | |
Collapse
|
5
|
Gress C, Vogel-Claussen J, Badorrek P, Müller M, Hohl K, Konietzke M, Litzenburger T, Seibold W, Gupta A, Hohlfeld JM. The effect of bradykinin 1 receptor antagonist BI 1026706 on pulmonary inflammation after segmental lipopolysaccharide challenge in healthy smokers. Pulm Pharmacol Ther 2023; 82:102246. [PMID: 37562641 DOI: 10.1016/j.pupt.2023.102246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/25/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Bradykinin 1 receptor (B1R) signalling pathways may be involved in the inflammatory pathophysiology of chronic obstructive pulmonary disease (COPD). B1R signalling is induced by inflammatory stimuli or tissue injury and leads to activation and increased migration of pro-inflammatory cells. Lipopolysaccharide (LPS) lung challenge in man is an experimental method of exploring inflammation in the lung whereby interference in these pathways can help to assess pharmacologic interventions in COPD. BI 1026706, a potent B1R antagonist, was hypothesized to reduce the inflammatory activity after segmental lipopolysaccharide (LPS) challenge in humans due to decreased pulmonary cell influx. METHODS In a monocentric, randomized, double-blind, placebo-controlled, parallel-group, phase I trial, 57 healthy, smoking subjects were treated for 28 days with either oral BI 1026706 100 mg bid or placebo. At day 21, turbo-inversion recovery magnitude magnetic resonance imaging (TIRM MRI) was performed. On the last day of treatment, pre-challenge bronchoalveolar lavage fluid (BAL) and biopsies were sampled, followed by segmental LPS challenge (40 endotoxin units/kg body weight) and saline control instillation in different lung lobes. Twenty-four hours later, TIRM MRI was performed, then BAL and biopsies were collected from the challenged segments. In BAL samples, cells were differentiated for neutrophil numbers as the primary endpoint. Other endpoints included assessment of safety, biomarkers in BAL (e.g. interleukin-8 [IL-8], albumin and total protein), B1R expression in lung biopsies and TIRM score by MRI as a measure for the extent of pulmonary oedema. RESULTS After LPS, but not after saline, high numbers of inflammatory cells, predominantly neutrophils were observed in the airways. IL-8, albumin and total protein were also increased in BAL samples after LPS challenge as compared with saline control. There were no significant differences in cells or other biomarkers from BAL in volunteers treated with BI 1026706 compared with those treated with placebo. Unexpectedly, neutrophil numbers in BAL were 30% higher and MRI-derived extent of oedema was significantly higher with BI 1026706 treatment compared with placebo, 24 h after LPS challenge. Adverse events were mainly mild to moderate and not different between treatment groups. CONCLUSIONS Treatment with BI 1026706 for four weeks was safe and well-tolerated in healthy smoking subjects. BI 1026706 100 mg bid did not provide evidence for anti-inflammatory effects in the human bronchial LPS challenge model. TRIAL REGISTRATION The study was registered on January 14, 2016 at ClinicalTrials.gov (NCT02657408).
Collapse
Affiliation(s)
- Christina Gress
- Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany; German Center for Lung Research (BREATH), Hannover, Germany
| | - Jens Vogel-Claussen
- German Center for Lung Research (BREATH), Hannover, Germany; Department of Diagnostic and Interventional Radiology, Hannover Medical School, Germany
| | - Philipp Badorrek
- Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Meike Müller
- Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany; German Center for Lung Research (BREATH), Hannover, Germany
| | - Kathrin Hohl
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | | | | | - Abhya Gupta
- Boehringer Ingelheim International GmbH, Biberach, Germany
| | - Jens M Hohlfeld
- Clinical Airway Research, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany; German Center for Lung Research (BREATH), Hannover, Germany; Department of Respiratory Medicine, Hannover Medical School, Germany.
| |
Collapse
|
6
|
Ware LB, Soleymanlou N, McAuley DF, Estrada V, Diaz GA, Lacamera P, Kaste R, Choi W, Gupta A, Welte T. TRPC6 inhibitor (BI 764198) to reduce risk and severity of ARDS due to COVID-19: a phase II randomised controlled trial. Thorax 2023; 78:816-824. [PMID: 37024277 PMCID: PMC10359525 DOI: 10.1136/thorax-2022-219668] [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: 09/23/2022] [Accepted: 01/31/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND Despite the availability of COVID-19 vaccinations, there remains a need to investigate treatments to reduce the risk or severity of potentially fatal complications of COVID-19, such as acute respiratory distress syndrome (ARDS). This study evaluated the efficacy and safety of the transient receptor potential channel C6 (TRPC6) inhibitor, BI 764198, in reducing the risk and/or severity of ARDS in patients hospitalised for COVID-19 and requiring non-invasive, supplemental oxygen support (oxygen by mask or nasal prongs, oxygen by non-invasive ventilation or high-flow nasal oxygen). METHODS Multicentre, double-blind, randomised phase II trial comparing once-daily oral BI 764198 (n=65) with placebo (n=64) for 28 days (+2-month follow-up). PRIMARY ENDPOINT proportion of patients alive and free of mechanical ventilation at day 29. Secondary endpoints: proportion of patients alive and discharged without oxygen (day 29); occurrence of either in-hospital mortality, intensive care unit admission or mechanical ventilation (day 29); time to first response (clinical improvement/recovery); ventilator-free days (day 29); and mortality (days 15, 29, 60 and 90). RESULTS No difference was observed for the primary endpoint: BI 764198 (83.1%) versus placebo (87.5%) (estimated risk difference -5.39%; 95% CI -16.08 to 5.30; p=0.323). For secondary endpoints, a longer time to first response (rate ratio 0.67; 95% CI 0.46 to 0.99; p=0.045) and longer hospitalisation (+3.41 days; 95% CI 0.49 to 6.34; p=0.023) for BI 764198 versus placebo was observed; no other significant differences were observed. On-treatment adverse events were similar between trial arms and more fatal events were reported for BI 764198 (n=7) versus placebo (n=2). Treatment was stopped early based on an interim observation of a lack of efficacy and an imbalance of fatal events (Data Monitoring Committee recommendation). CONCLUSIONS TRPC6 inhibition was not effective in reducing the risk and/or severity of ARDS in patients with COVID-19 requiring non-invasive, supplemental oxygen support. TRIAL REGISTRATION NUMBER NCT04604184.
Collapse
Affiliation(s)
- Lorraine B Ware
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Nima Soleymanlou
- TA Cardio-Metabolism & Respiratory, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut, USA
| | - Danny Francis McAuley
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast Wellcome-Wolfson Institute for Experimental Medicine, Belfast, UK
| | - Vicente Estrada
- Hospital Clínico San Carlos, IdISSC; CIBERINFE, Madrid, Spain
| | - George A Diaz
- Section of Infectious Diseases, Providence Regional Medical Center Everett, Everett, Washington, USA
| | - Peter Lacamera
- Division of Pulmonary and Critical Care Medicine, St Elizabeth's Medical Center, Boston, Massachusetts, USA
| | - Renee Kaste
- TA Cardio-Metabolism & Respiratory, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut, USA
| | - Wansuk Choi
- TA Cardio-Metabolism & Respiratory, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut, USA
| | - Abhya Gupta
- TA Inflammation Medicine, Boehringer Ingelheim International GmbH, Biberach an der Riss, Germany
| | - Tobias Welte
- Department of Pneumology, Hannover Medical School, Hannover, Niedersachsen, Germany
| |
Collapse
|
7
|
Rampa DR, Feng H, Allur-Subramaniyan S, Shim K, Pekcec A, Lee D, Doods H, Wu D. Kinin B1 receptor blockade attenuates hepatic fibrosis and portal hypertension in chronic liver diseases in mice. J Transl Med 2022; 20:590. [PMID: 36514072 PMCID: PMC9746183 DOI: 10.1186/s12967-022-03808-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/03/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND AIMS Kinin B1 receptors (B1Rs) are implicated in the pathogenesis of fibrosis. This study examined the anti-fibrotic effects of B1R blockade with BI 113823 in two established mouse models of hepatic fibrosis induced by intraperitoneal carbon tetrachloride (CCl4) injection or bile duct ligation (BDL). The mechanisms underlying the protection afforded by B1R inhibition were examined using human peripheral blood cells and LX2 human hepatic stellate cells (HSCs). METHODS Fibrotic liver diseases were induced in mice by intraperitoneal carbon tetrachloride (CCl4) injection for 6 weeks, and by bile duct ligation (BDL) for 3 weeks, respectively. Mice received daily treatment of vehicle or BI 113823 (B1R antagonist) from onset of the experiment until the end of the study. RESULTS B1Rs were strongly induced in fibrotic mouse liver. BI 113823 significantly attenuated liver fibrosis and portal hypertension (PH), and improved survival in both CCl4 and BDL mice. BI 113823 significantly reduced the expression of fibrotic proteins α-SMA, collagens 1, 3, 4, and profibrotic growth factors PDGF, TGFβ, CTGF, VEGF, proliferating cell nuclear antigen; and reduced hepatic Akt phosphorylation in CCl4- and BDL-induced liver fibrosis. BI 113823 also reduced expression of Cytokines IL-1, IL-6; chemokines MCP-1, MCP-3 and infiltration of inflammatory cells; and inhibited human monocyte and neutrophil activation, transmigration, TNF-α & MPO production in vitro. BI 113823 inhibited TGF-β and B1R agonist-stimulated human-HSC activation, contraction, proliferation, migration and fibrosis protein expression, and inhibited activation of PI3K/Akt signalling pathway. CONCLUSIONS B1Rs merits consideration as a novel therapeutic target for chronic liver fibrosis and PH.
Collapse
Affiliation(s)
- Dileep Reddy Rampa
- grid.411545.00000 0004 0470 4320Department of Bio-Nanotechnology and Bio-Convergence Engineering, Jeonbuk National University, Jeonju, South Korea
| | - Huiying Feng
- grid.411545.00000 0004 0470 4320Department of Bio-Nanotechnology and Bio-Convergence Engineering, Jeonbuk National University, Jeonju, South Korea
| | - Sivakumar Allur-Subramaniyan
- grid.411545.00000 0004 0470 4320Department of Animal Biotechnology & Agricultural Convergence Technology, Jeonbuk National University, Jeonju, South Korea
| | - Kwanseob Shim
- grid.411545.00000 0004 0470 4320Department of Animal Biotechnology & Agricultural Convergence Technology, Jeonbuk National University, Jeonju, South Korea
| | - Anton Pekcec
- grid.420061.10000 0001 2171 7500Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Dongwon Lee
- grid.411545.00000 0004 0470 4320Department of Bio-Nanotechnology and Bio-Convergence Engineering, Jeonbuk National University, Jeonju, South Korea
| | - Henri Doods
- grid.420061.10000 0001 2171 7500Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Dongmei Wu
- grid.411545.00000 0004 0470 4320Department of Bio-Nanotechnology and Bio-Convergence Engineering, Jeonbuk National University, Jeonju, South Korea ,grid.410396.90000 0004 0430 4458Department of Research, Mount Sinai Medical Center, Miami Beach, FL USA
| |
Collapse
|
8
|
Bradykinin-target therapies in SARS-CoV-2 infection: current evidence and perspectives. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:275-283. [PMID: 35089406 PMCID: PMC8795307 DOI: 10.1007/s00210-022-02206-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 01/17/2022] [Indexed: 12/26/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a potentially fatal disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that preferentially infects the respiratory tract. Bradykinin (BK) is a hypotensive substance that recently emerged as one of the mechanisms to explain COVID-19-related complications. Concerning this, in this review, we try to address the complex link between BK and pathophysiology of COVID-19, investigating the role of this peptide as a potential target for pharmacological modulation in the management of SARS-CoV-2. The pathology of COVID-19 may be more a result of the BK storm than the cytokine storm, and which BK imbalance is a relevant factor in the respiratory disorders caused by SARS-CoV-2 infection. Regarding this, an interesting point of intervention for this disease is to modulate BK signaling. Some drugs, such as icatibant, ecallantide, and noscapine, and even a human monoclonal antibody, lanadelumab, have been studied for their potential utility in COVID-19 by modulating BK signaling. The interaction of the BK pathway and the involvement of cytokines such as IL-6 and IL1 may be key to the use of blockers, even if only as adjuvants. In fact, reduction of BK, mainly DABK, is considered a relevant strategy to improve clinical conditions of COVID-19 patients. In this context, despite the current unproven clinical efficacy, drugs repurposing that block B1 or B2 receptor activation have gained prominence for the treatment of COVID-19 in the world.
Collapse
|
9
|
Kiatamornrak P, Boobphahom S, Lertussavavivat T, Rattanawaleedirojn P, Chailapakul O, Rodthongkum N, Srisawat N. A portable blood lactate sensor with a non-immobilized enzyme for early sepsis diagnosis. Analyst 2022; 147:2819-2827. [DOI: 10.1039/d2an00218c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Early determination of blood lactate levels may accelerate the detection of sepsis, one of the most time-sensitive illnesses.
Collapse
Affiliation(s)
- Patcharakorn Kiatamornrak
- Excellence Center for Critical Care Nephrology, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Pathumwan, Bangkok 10330, Thailand
- Center of Excellence in Critical Care Nephrology, Chulalongkorn University, Thailand
| | - Siraprapa Boobphahom
- Metallurgy and Materials Science Research Institute, Chulalongkorn University, Soi Chula12, Phayathai Road, Pathumwan, Bangkok 10330, Thailand
| | - Tanat Lertussavavivat
- Excellence Center for Critical Care Nephrology, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Pathumwan, Bangkok 10330, Thailand
- Center of Excellence in Critical Care Nephrology, Chulalongkorn University, Thailand
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | - Pranee Rattanawaleedirojn
- Metallurgy and Materials Science Research Institute, Chulalongkorn University, Soi Chula12, Phayathai Road, Pathumwan, Bangkok 10330, Thailand
| | - Orawon Chailapakul
- Electrochemistry and Optical Spectroscopy Center of Excellence, Department of Chemistry, Faculty of Science, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | - Nadnudda Rodthongkum
- Metallurgy and Materials Science Research Institute, Chulalongkorn University, Soi Chula12, Phayathai Road, Pathumwan, Bangkok 10330, Thailand
| | - Nattachai Srisawat
- Excellence Center for Critical Care Nephrology, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Pathumwan, Bangkok 10330, Thailand
- Center of Excellence in Critical Care Nephrology, Chulalongkorn University, Thailand
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
- Academy of Science, Royal Society of Thailand, Bangkok, Thailand
| |
Collapse
|
10
|
Oliveira LCG, Cruz NAN, Ricelli B, Tedesco-Silva H, Medina-Pestana JO, Casarini DE. Interactions amongst inflammation, renin-angiotensin-aldosterone and kallikrein-kinin systems: suggestive approaches for COVID-19 therapy. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20200181. [PMID: 34925477 PMCID: PMC8651214 DOI: 10.1590/1678-9199-jvatitd-2020-0181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/24/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a rapid-spread infectious disease caused by the SARS-CoV-2 virus, which can culminate in the renin-angiotensin-aldosterone (RAAS) and kallikrein-kinin (KKS) systems imbalance, and in serious consequences for infected patients. This scoping review of published research exploring the RAAS and KKS was undertaken in order to trace the history of the discovery of both systems and their multiple interactions, discuss some aspects of the viral-cell interaction, including inflammation and the system imbalance triggered by SARS-CoV-2 infection, and their consequent disorders. Furthermore, we correlate the effects of continued use of the RAAS blockers in chronic diseases therapies with the virulence and physiopathology of COVID-19. We also approach the RAAS and KKS-related proposed potential therapies for treatment of COVID-19. In this way, we reinforce the importance of exploring both systems and the application of their components or their blockers in the treatment of coronavirus disease.
Collapse
Affiliation(s)
| | | | - Bruna Ricelli
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, SP, Brazil
| | - Helio Tedesco-Silva
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, SP, Brazil
| | - José Osmar Medina-Pestana
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, SP, Brazil
| | - Dulce Elena Casarini
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, SP, Brazil
| |
Collapse
|
11
|
Rampa DR, Murugesan P, Chao H, Feng H, Dai W, Lee D, Pekcec A, Doods H, Wu D. Reversal of pulmonary arterial hypertension and neointimal formation by kinin B1 receptor blockade. Respir Res 2021; 22:281. [PMID: 34717626 PMCID: PMC8557528 DOI: 10.1186/s12931-021-01875-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 10/19/2021] [Indexed: 11/15/2022] Open
Abstract
Background This study examined whether BI113823, a novel selective kinin B1 receptor antagonist can reverse established pulmonary arterial hypertension (PAH), prevent right heart failure and death, which is critical for clinical translation. Methods Left pneumonectomized male Wistar rats were injected with monocrotaline to induce PAH. Three weeks later, when PAH was well established, the rats received daily treatment of BI113823 or vehicle for 3 weeks. Results Treatment with BI113823 from day 21 to day 42 after monocrotaline injection reversed established PAH as shown by normalized values of mean pulmonary arterial pressure (mPAP). BI113823 therapy reversed pulmonary vascular remodeling, pulmonary arterial neointimal formation, and heart and lung fibrosis, reduced right ventricular pressure, right heart hypertrophy, improved cardiac output, and prevented right heart failure and death. Treatment with BI113823 reduced TNF-α and IL-1β, and macrophages recruitment in bronchoalveolar lavage, reduced CD-68 positive macrophages and expression of proliferating cell nuclear antigen (PCNA) in the perivascular areas, and reduced expression of iNOS, B1 receptors, matrix metalloproteinase (MMP)-2 and MMP-9 proteins, and the phosphorylation of ERK1/2 and AKT in lung. Treatment with BI113823 reduced mRNA expression of ANP, BNP, βMHC, CGTF, collange-I and IV in right heart, compared to vehicle treated controls. In human monocytes cultures, BI113823 reduced LPS-induced TNF-α production, MMP-2 and MMP-9 expression, and reduced TNF-α-induced monocyte migration. Conclusions We conclude that BI113823 reverses preexisting severe experimental pulmonary hypertension via inhibition of macrophage infiltration, cytokine production, as well as down regulation of matrix metalloproteinase proteins.
Collapse
Affiliation(s)
- Dileep Reddy Rampa
- Department of Bio-Nanotechnology and Bio-Convergence Engineering, Chonbuk National University, Jeonju, South Korea
| | - Priya Murugesan
- Department of Bio-Nanotechnology and Bio-Convergence Engineering, Chonbuk National University, Jeonju, South Korea
| | - Honglu Chao
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huiying Feng
- Department of Bio-Nanotechnology and Bio-Convergence Engineering, Chonbuk National University, Jeonju, South Korea.,Department of Research, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Wenxin Dai
- Department of Bio-Nanotechnology and Bio-Convergence Engineering, Chonbuk National University, Jeonju, South Korea
| | - Dongwon Lee
- Department of Bio-Nanotechnology and Bio-Convergence Engineering, Chonbuk National University, Jeonju, South Korea
| | - Anton Pekcec
- Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Henri Doods
- Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Dongmei Wu
- Department of Bio-Nanotechnology and Bio-Convergence Engineering, Chonbuk National University, Jeonju, South Korea. .,Department of Research, Mount Sinai Medical Center, Miami Beach, FL, USA.
| |
Collapse
|
12
|
Inhibition of microsomal prostaglandin E synthase-1 ameliorates acute lung injury in mice. J Transl Med 2021; 19:340. [PMID: 34372885 PMCID: PMC8351447 DOI: 10.1186/s12967-021-03016-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 07/29/2021] [Indexed: 11/23/2022] Open
Abstract
Background To examine the effects of BI 1029539 (GS-248), a novel selective human microsomal prostaglandin E synthase-1 (mPGES-1) inhibitor, in experimental models of acute lung injury (ALI) and sepsis in transgenic mice constitutively expressing the mPGES1 (Ptges) humanized allele. Methods Series 1: Lipopolysaccharide (LPS)-induced ALI. Mice were randomized to receive vehicle, BI 1029539, or celecoxib. Series 2: Cecal ligation and puncture-induced sepsis. Mice were randomized to receive vehicle or BI 1029539. Results Series 1: BI 1029539 or celecoxib reduced LPS-induced lung injury, with reduction in neutrophil influx, protein content, TNF-ɑ, IL-1β and PGE2 levels in bronchoalveolar lavage (BAL), myeloperoxidase activity, expression of mPGES-1, cyclooxygenase (COX)-2 and intracellular adhesion molecule in lung tissue compared with vehicle-treated mice. Notably, prostacyclin (PGI2) BAL concentration was only lowered in celecoxib-treated mice. Series 2: BI 1029539 significantly reduced sepsis-induced BAL inflammatory cell recruitment, lung injury score and lung expression of mPGES-1 and inducible nitric oxide synthase. Treatment with BI 1029539 also significantly prolonged survival of mice with severe sepsis. Anti-inflammatory and anti-migratory effect of BI 1029539 was confirmed in peripheral blood leukocytes from healthy volunteers. Conclusions BI 1029539 ameliorates leukocyte infiltration and lung injury resulting from both endotoxin-induced and sepsis-induced lung injury. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03016-9.
Collapse
|
13
|
Ayola-Serrano NC, Roy N, Fathah Z, Anwar MM, Singh B, Ammar N, Sah R, Elba A, Utt RS, Pecho-Silva S, Rodriguez-Morales AJ, Dhama K, Quraishi S. The role of 5-lipoxygenase in the pathophysiology of COVID-19 and its therapeutic implications. Inflamm Res 2021; 70:877-889. [PMID: 34086061 PMCID: PMC8176665 DOI: 10.1007/s00011-021-01473-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/21/2021] [Accepted: 05/15/2021] [Indexed: 12/14/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, known as coronavirus disease 2019 (COVID-19) causes cytokine release syndrome (CRS), leading to acute respiratory distress syndrome (ARDS), acute kidney and cardiac injury, liver dysfunction, and multiorgan failure. Although several studies have discussed the role of 5-lipoxygenase (5-LOX) in viral infections, such as influenzae and SARS, it remains unexplored in the pathophysiology of COVID-19. 5-LOX acts on free arachidonic acid (AA) to form proinflammatory leukotrienes (LTs). Of note, numerous cells involved with COVID-19 (e.g., inflammatory and smooth muscle cells, platelets, and vascular endothelium) widely express leukotriene receptors. Moreover, 5-LOX metabolites induce the release of cytokines (e.g., tumour necrosis factor-α [TNF-α], interleukin-1α [IL-1α], and interleukin-1β [IL-1β]) and express tissue factor on cell membranes and activate plasmin. Since macrophages, monocytes, neutrophils, and eosinophils can express lipoxygenases, activation of 5-LOX and the subsequent release of LTs may contribute to the severity of COVID-19. This review sheds light on the potential implications of 5-LOX in SARS-CoV-2-mediated infection and the anticipated therapeutic role of 5-LOX inhibitors.
Collapse
Affiliation(s)
| | - Namrata Roy
- SRM University, SRM Nagar, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India.
| | | | - Mohammed Moustapha Anwar
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, Egypt
| | | | - Nour Ammar
- Department of Pediatric Dentistry and Dental Public Health, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| | - Ranjit Sah
- Department of Microbiology, Institute of Medicine, Tribhuvan University Teaching Hospital, Kathmandu, Nepal
| | - Areej Elba
- Department of Pediatric Dentistry and Dental Public Health, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| | - Rawan Sobhi Utt
- Faculty of Medicine, Al Quds University, Jerusalem, Palestine
| | - Samuel Pecho-Silva
- Master in Clinical Epidemiology and Biostatistics, Universidad Científica del Sur, Lima, Peru
- Pneumology Service, Hospital Nacional Edgardo Rebagliati Martins, Lima, Peru
- Latin American Network of COVID-19 Research, Pereira, Colombia
| | - Alfonso J Rodriguez-Morales
- Master in Clinical Epidemiology and Biostatistics, Universidad Científica del Sur, Lima, Peru.
- Latin American Network of COVID-19 Research, Pereira, Colombia.
- Grupo de Investigacion Biomedicina, Faculty of Medicine, Fundacion Universitaria Autonoma de Las Americas, Pereira, Risaralda, Colombia.
- School of Medicine, Universidad Privada Franz Tamayo (UNIFRANZ), Cochabamba, Bolivia.
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243122, India
| | - Sadeq Quraishi
- Department of Anesthesiology & Perioperative Medicine - Tufts Medical Center, Tufts University School of Medicine, 800 Washington St, Ziskind 6038, Boston, MA, 02111, USA
| |
Collapse
|
14
|
Gouda AS, Adbelruhman FG, Elbendary RN, Alharbi FA, Alhamrani SQ, Mégarbane B. A comprehensive insight into the role of zinc deficiency in the renin-angiotensin and kinin-kallikrein system dysfunctions in COVID-19 patients. Saudi J Biol Sci 2021; 28:3540-3547. [PMID: 33746538 PMCID: PMC7962980 DOI: 10.1016/j.sjbs.2021.03.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Hypozincemia is prevalent in severe acute respiratory syndrome coronavirus-2 (SARS-COV-2)-infected patients and has been considered as a risk factor in severe coronavirus disease-2019 (COVID-19). Whereas zinc might affect SARS-COV-2 replication and cell entry, the link between zinc deficiency and COVID-19 severity could also be attributed to the effects of COVID-19 on the body metabolism and immune response. Zinc deficiency is more prevalent in the elderly and patients with underlying chronic diseases, with established deleterious consequences such as the increased risk of respiratory infection. We reviewed the expected effects of zinc deficiency on COVID-19-related pathophysiological mechanisms focusing on both the renin-angiotensin and kinin-kallikrein systems. Mechanisms and effects were extrapolated from the available scientific literature. Zinc deficiency alters angiotensin-converting enzyme-2 (ACE2) function, leading to the accumulation of angiotensin II, des-Arg9-bradykinin and Lys-des-Arg9-bradykinin, which results in an exaggerated pro-inflammatory response, vasoconstriction and pro-thrombotic effects. Additionally, zinc deficiency blocks the activation of the plasma contact system, a protease cascade initiated by factor VII activation. Suggested mechanisms include the inhibition of Factor XII activation and limitation of high-molecular-weight kininogen, prekallikrein and Factor XII to bind to endothelial cells. The subsequent accumulation of Factor XII and deficiency in bradykinin are responsible for increased production of inflammatory mediators and marked hypercoagulability, as typically observed in COVID-19 patients. To conclude, zinc deficiency may affect both the renin-angiotensin and kinin-kallikrein systems, leading to the exaggerated inflammatory manifestations characteristic of severe COVID-19.
Collapse
Affiliation(s)
- Ahmed S. Gouda
- National Egyptian Center for Toxicological Researches, Faculty of Medicine, Cairo University, Cairo, Egypt
- Poison Control and Forensic Chemistry Center, Northern Borders, Ministry of Health, Saudi Arabia
| | - Fatima G. Adbelruhman
- Department of Clinical Pathology, Alzahraa Hospital, Al-Azhar University, Cairo, Egypt
| | - Reham N. Elbendary
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Fadiyah Ahmed Alharbi
- Tabuk Poison Control and Forensic Medicinal Chemistry Center, Ministry of health, Saudi Arabia
| | - Sultan Qalit Alhamrani
- Tabuk Poison Control and Forensic Medicinal Chemistry Center, Ministry of health, Saudi Arabia
| | - Bruno Mégarbane
- Department of Medical and Toxicological Critical Care, Lariboisière Hospital, University of Paris, INSERM UMRS-1144, Paris, France
| |
Collapse
|
15
|
Scialo F, Daniele A, Amato F, Pastore L, Matera MG, Cazzola M, Castaldo G, Bianco A. ACE2: The Major Cell Entry Receptor for SARS-CoV-2. Lung 2020; 198:867-877. [PMID: 33170317 PMCID: PMC7653219 DOI: 10.1007/s00408-020-00408-4] [Citation(s) in RCA: 294] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023]
Abstract
Despite the unprecedented effort of the scientific community, the novel SARS-CoV-2 virus has infected more than 46 million people worldwide, killing over one million two hundred thousand. Understanding the mechanisms by which some individuals are more susceptible to SARS-CoV-2 infection and why a subgroup of them are prone to experience severe pneumonia, and death should lead to a better approach and more effective treatments for COVID-19. Here, we focus our attention on ACE2, a primary receptor of SARS-CoV-2. We will discuss its biology, tissue expression, and post-translational regulation that determine its potential to be employed by SARS-CoV-2 for cell entry. Particular attention will be given to how the ACE2 soluble form can have a great impact on disease progression and thus be used in a potential therapeutic strategy. Furthermore, we will discuss repercussions that SARS-CoV-2/ACE2 binding has on the renin–angiotensin system and beyond. Indeed, although mostly neglected, ACE2 can also act on [des-Arg 937]-bradykinin of the kinin–kallikrein system regulating coagulation and inflammation. Thorough comprehension of the role that ACE2 plays in different pathways will be the key to assess the impact that SARS-CoV-2/ACE2 binding has on organismal physiology and will help us to find better therapies and diagnostic tools.
Collapse
Affiliation(s)
- Filippo Scialo
- Dipartimento di Scienze Mediche Traslazionali, University of Campania "L. Vanvitelli", Naples, Italy
- CEINGE, Biotecnologie Avanzate, Naples, Italy
| | - Aurora Daniele
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, University of Campania "L. Vanvitelli", Naples, Italy
- CEINGE, Biotecnologie Avanzate, Naples, Italy
| | - Felice Amato
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Di Napoli Federico II, Naples, Italy
- CEINGE, Biotecnologie Avanzate, Naples, Italy
| | - Lucio Pastore
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Di Napoli Federico II, Naples, Italy
- CEINGE, Biotecnologie Avanzate, Naples, Italy
| | - Maria Gabriella Matera
- Dipartimento di Medicina Sperimentale, University of Campania "L. Vanvitelli", Naples, Italy
| | - Mario Cazzola
- Dipartimento di Medicina Sperimentale, University of Rome "Tor Vergata", Rome, Italy.
| | - Giuseppe Castaldo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Di Napoli Federico II, Naples, Italy
- CEINGE, Biotecnologie Avanzate, Naples, Italy
| | - Andrea Bianco
- Dipartimento di Scienze Mediche Traslazionali, University of Campania "L. Vanvitelli", Naples, Italy
| |
Collapse
|
16
|
Sharifkashani S, Bafrani MA, Khaboushan AS, Pirzadeh M, Kheirandish A, Yavarpour Bali H, Hessami A, Saghazadeh A, Rezaei N. Angiotensin-converting enzyme 2 (ACE2) receptor and SARS-CoV-2: Potential therapeutic targeting. Eur J Pharmacol 2020; 884:173455. [PMID: 32745604 PMCID: PMC7834210 DOI: 10.1016/j.ejphar.2020.173455] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/30/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a beta coronavirus that uses the human angiotensin-converting enzyme 2 (ACE2) receptor as a point of entry. The present review discusses the origin and structure of the virus and its mechanism of cell entry followed by the therapeutic potentials of strategies directed towards SARS-CoV2-ACE2 binding, the renin-angiotensin system, and the kinin-kallikrein system. SARS-CoV2-ACE2 binding-directed approaches mainly consist of targeting receptor binding domain, ACE2 blockers, soluble ACE2, and host protease inhibitors. In conclusion, blocking or manipulating the SARS-CoV2-ACE2 binding interface perhaps offers the best tactic against the virus that should be treated as a fundamental subject of future research.
Collapse
Affiliation(s)
- Sourena Sharifkashani
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Melika Arab Bafrani
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Soltani Khaboushan
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Pirzadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Ali Kheirandish
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Student Research Committee, School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hanie Yavarpour Bali
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Amirhossein Hessami
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amene Saghazadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
17
|
Dagnino APA, Campos MM, Silva RBM. Kinins and Their Receptors in Infectious Diseases. Pharmaceuticals (Basel) 2020; 13:ph13090215. [PMID: 32867272 PMCID: PMC7558425 DOI: 10.3390/ph13090215] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 01/08/2023] Open
Abstract
Kinins and their receptors have been implicated in a series of pathological alterations, representing attractive pharmacological targets for several diseases. The present review article aims to discuss the role of the kinin system in infectious diseases. Literature data provides compelling evidence about the participation of kinins in infections caused by diverse agents, including viral, bacterial, fungal, protozoan, and helminth-related ills. It is tempting to propose that modulation of kinin actions and production might be an adjuvant strategy for management of infection-related complications.
Collapse
|
18
|
Gouda AS, Mégarbane B. Snake venom-derived bradykinin-potentiating peptides: A promising therapy for COVID-19? Drug Dev Res 2020; 82:38-48. [PMID: 32761647 PMCID: PMC7436322 DOI: 10.1002/ddr.21732] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/23/2022]
Abstract
The severe acute respiratory syndrome coronavirus‐2 (SARS‐COV‐2), a novel coronavirus responsible for the recent infectious pandemic, is known to downregulate angiotensin‐converting enzyme‐2 (ACE2). Most current investigations focused on SARS‐COV‐2‐related effects on the renin–angiotensin system and especially the resultant increase in angiotensin II, neglecting its effects on the kinin–kallikrein system. SARS‐COV‐2‐induced ACE2 inhibition leads to the augmentation of bradykinin 1‐receptor effects, as ACE2 inactivates des‐Arg9‐bradykinin, a bradykinin metabolite. SARS‐COV‐2 also decreases bradykinin 2‐receptor effects as it affects bradykinin synthesis by inhibiting cathepsin L, a kininogenase present at the site of infection and involved in bradykinin production. The physiologies of both the renin–angiotensin and kinin–kallikrein system are functionally related suggesting that any intervention aiming to treat SARS‐COV‐2‐infected patients by triggering one system but ignoring the other may not be adequately effective. Interestingly, the snake‐derived bradykinin‐potentiating peptide (BPP‐10c) acts on both systems. BPP‐10c strongly decreases angiotensin II by inhibiting ACE, increasing bradykinin‐related effects on the bradykinin 2‐receptor and increasing nitric oxide‐mediated effects. Based on a narrative review of the literature, we suggest that BPP‐10c could be an optimally effective option to consider when aiming at developing an anti‐SARS‐COV‐2 drug.
Collapse
Affiliation(s)
- Ahmed S Gouda
- National Egyptian Center for Toxicological Researches, Faculty of Medicine, University of Cairo, Cairo, Egypt
| | - Bruno Mégarbane
- Department of Medical and Toxicological Critical Care, Lariboisière Hospital, University of Paris, INSERM UMRS-1144, Paris, France
| |
Collapse
|
19
|
Gregnani MF, Hungaro TG, Martins-Silva L, Bader M, Araujo RC. Bradykinin B2 Receptor Signaling Increases Glucose Uptake and Oxidation: Evidence and Open Questions. Front Pharmacol 2020; 11:1162. [PMID: 32848770 PMCID: PMC7417865 DOI: 10.3389/fphar.2020.01162] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/17/2020] [Indexed: 12/21/2022] Open
Abstract
The Kinin B2 receptor (B2R) is classically involved in vasodilation and inflammatory responses. However, through the observation of hypoglycemic effects of Angiotensin-I-Converting Enzyme (ACE) inhibitors, this protein has been related to metabolic glucose modulation in physiological and pathophysiological contexts. Although several studies have evaluated this matter, the different methodologies and models employed, combined with the distinct target organs, results in a challenge to summarize and apply the knowledge in this field. Therefore, this review aims to compile human and animal data in order to provide a big picture about what is already known regarding B2R and glucose metabolism, as well to suggest pending investigation issues aiming at evaluating the role of B2R in relation to glucose metabolism in homeostatic situations and metabolic disturbances. The data indicate that B2R signaling is involved mainly in glucose uptake in skeletal muscle and adipose tissue, acting as a synergic player beside insulin. However, most data indicate that B2R induces increased glucose oxidation, instead of storage, via activation of a broad signaling cascade involving Nitric Oxide (NO) and cyclic-GMP dependent protein kinase (PKG). Additionally, we highlight that this modulation is impaired in metabolic disturbances such as diabetes and obesity, and we provide a hypothetic mechanism to explain this blockade in light of literature data provided for this review, as well as other authors.
Collapse
Affiliation(s)
- Marcos Fernandes Gregnani
- Laboratory of Genetic and Metabolism of Exercise, Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil.,Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Talita G Hungaro
- Laboratory of Genetic and Metabolism of Exercise, Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil.,Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | | | - Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany.,Institute for Biology, University of Lübeck, Lübeck, Germany.,Charité University Medicine, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Ronaldo C Araujo
- Laboratory of Genetic and Metabolism of Exercise, Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Nicolau LAD, Magalhães PJC, Vale ML. What would Sérgio Ferreira say to your physician in this war against COVID-19: How about kallikrein/kinin system? Med Hypotheses 2020; 143:109886. [PMID: 32504925 PMCID: PMC7261103 DOI: 10.1016/j.mehy.2020.109886] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/25/2020] [Indexed: 12/29/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease with fast spreading all over the world caused by the SARS-CoV-2 virus which can culminate in a severe acute respiratory syndrome by the injury caused in the lungs. However, other organs can be also damaged. SARS-CoV-2 enter into the host cells using the angiotensin-converting enzyme 2 (ACE2) as receptor, like its ancestor SARS-CoV. ACE2 is then downregulated in lung tissues with augmented serum levels of ACE2 in SARS-CoV-2 patients. Interestingly, ACE2+ organs reveal the symptomatic repercussions, which are signals of the infection such as dry cough, shortness of breath, heart failure, liver and kidney damage, anosmia or hyposmia, and diarrhea. ACE2 exerts a chief role in the renin-angiotensin system (RAS) by converting angiotensin II to angiotensin-(1-7) that activates Mas receptor, inhibits ACE1, and modulates bradykinin (BK) receptor sensitivity, especially the BK type 2 receptor (BKB2R). ACE2 also hydrolizes des-Arg9-bradykinin (DABK), an active BK metabolite, agonist at BK type 1 receptors (BKB1R), which is upregulated by inflammation. In this opinion article, we conjecture a dialogue by the figure of Sérgio Ferreira which brought together basic science of classical pharmacology and clinical repercussions in COVID-19, then we propose that in the course of SARS-CoV-2 infection: i) downregulation of ACE2 impairs the angiotensin II and DABK inactivation; ii) BK and its metabolite DABK seems to be in elevated levels in tissues by interferences in kallikrein/kinin system; iii) BK1 receptor contributes to the outbreak and maintenance of the inflammatory response; iv) kallikrein/kinin system crosstalks to RAS and coagulation system, linking inflammation to thrombosis and organ injury. We hypothesize that targeting the kallikrein/kinin system and BKB1R pathway may be beneficial in SARS-CoV-2 infection, especially on early stages. This route of inference should be experimentally verified by SARS-CoV-2 infected mice.
Collapse
Affiliation(s)
- Lucas A D Nicolau
- Biotechnology and Biodiversity Center Research, Federal University of Parnaíba Delta, Parnaíba, Brazil
| | - Pedro J C Magalhães
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Mariana L Vale
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil.
| |
Collapse
|
21
|
Roche JA, Roche R. A hypothesized role for dysregulated bradykinin signaling in COVID-19 respiratory complications. FASEB J 2020; 34:7265-7269. [PMID: 32359101 PMCID: PMC7267506 DOI: 10.1096/fj.202000967] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 01/13/2023]
Abstract
As of April 20, 2020, over time, the COVID-19 pandemic has resulted in 157 970 deaths out of 2 319 066 confirmed cases, at a Case Fatality Rate of ~6.8%. With the pandemic rapidly spreading, and health delivery systems being overwhelmed, it is imperative that safe and effective pharmacotherapeutic strategies are rapidly explored to improve survival. In this paper, we use established and emerging evidence to propose a testable hypothesis that, a vicious positive feedback loop of des-Arg(9)-bradykinin- and bradykinin-mediated inflammation → injury → inflammation, likely precipitates life threatening respiratory complications in COVID-19. Through our hypothesis, we make the prediction that the FDA-approved molecule, icatibant, might be able to interrupt this feedback loop and, thereby, improve the clinical outcomes. This hypothesis could lead to basic, translational, and clinical studies aimed at reducing COVID-19 morbidity and mortality.
Collapse
Affiliation(s)
- Joseph A Roche
- Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Renuka Roche
- College of Health and Human Services, Eastern Michigan University, Ypsilanti, MI, USA
| |
Collapse
|
22
|
Ruiz S, Vardon-Bounes F, Buléon M, Guilbeau-Frugier C, Séguelas MH, Conil JM, Girolami JP, Tack I, Minville V. Kinin B1 receptor: a potential therapeutic target in sepsis-induced vascular hyperpermeability. J Transl Med 2020; 18:174. [PMID: 32306971 PMCID: PMC7168845 DOI: 10.1186/s12967-020-02342-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/10/2020] [Indexed: 12/14/2022] Open
Abstract
Background In sepsis, the endothelial barrier becomes incompetent, with the leaking of plasma into interstitial tissues. VE-cadherin, an adherens junction protein, is the gatekeeper of endothelial cohesion. Kinins, released during sepsis, induce vascular leakage and vasodilation. They act via two G-protein coupled receptors: B1 (B1R) and B2 (B2R). B1R is inducible in the presence of pro-inflammatory cytokines, endotoxins or after tissue injury. It acts at a later stage of sepsis and elicits a sustained inflammatory response. The aim of our study was to investigate the relationships between B1R and VE-cadherin destabilization in vivo in a later phase of sepsis. Methods Experimental, prospective study in a university research laboratory. We used a polymicrobial model of septic shock by cecal ligation and puncture in C57BL6 male mice or C57BL6 male mice that received a specific B1R antagonist (R-954). We studied the influence of B1R on sepsis-induced vascular permeability 30 h after surgery for several organs, and VE-cadherin expression in the lung and kidneys by injecting R-954 just before surgery. The 96-h survival was determined in mice without treatment or in animals receiving R-954 as a “prophylactic” regimen (a subcutaneous injection of 200 µg/kg, prior to CLP and 24 h after CLP), or as a “curative” regimen (injection of 100 µg/kg at H6, H24 and H48 post-surgery). Results B1R inactivation helps to maintain MAP above 65 mmHg but induces different permeability profiles depending on whether or not organ perfusion is autoregulated. In our model, VE-cadherin was destabilized in vivo during septic shock. At a late stage of sepsis, the B1R blockade reduced the VE-cadherin disruption by limiting eNOS activation. The survival rate for mice that received R-954 after sepsis induction was higher than in animals that received an antagonist as a prophylactic treatment. Conclusions B1R antagonizing reduced mortality in our model of murine septic shock by limiting the vascular permeability induced by VE-cadherin destabilization through maintenance of the macrohemodynamics, consequently limiting organ dysfunctions.
Collapse
Affiliation(s)
- Stéphanie Ruiz
- Department of Anesthesiology and Intensive Care, Rangueil Hospital-University Hospital of Toulouse, 1 Avenue du Professeur Jean Poulhès TSA 50032, 31059, Toulouse Cedex 9, France. .,Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France.
| | - Fanny Vardon-Bounes
- Department of Anesthesiology and Intensive Care, Rangueil Hospital-University Hospital of Toulouse, 1 Avenue du Professeur Jean Poulhès TSA 50032, 31059, Toulouse Cedex 9, France.,Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France
| | - Marie Buléon
- Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France
| | - Céline Guilbeau-Frugier
- Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France.,Department of Forensic Medicine, Rangueil Hospital-University Hospital of Toulouse, 1 Avenue du Professeur Jean Poulhès TSA 50032, 31059, Toulouse Cedex 9, France.,Biological Electron Microscopy Center, Rangueil Faculty of Medicine, Toulouse University, Toulouse, France
| | - Marie-Hélène Séguelas
- Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France
| | - Jean-Marie Conil
- Department of Anesthesiology and Intensive Care, Rangueil Hospital-University Hospital of Toulouse, 1 Avenue du Professeur Jean Poulhès TSA 50032, 31059, Toulouse Cedex 9, France
| | - Jean-Pierre Girolami
- Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France
| | - Ivan Tack
- Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France.,Department of Physiology, Rangueil Hospital-University Hospital of Toulouse, 1 Avenue du Professeur Jean Poulhès TSA 50032, 31059, Toulouse Cedex 9, France
| | - Vincent Minville
- Department of Anesthesiology and Intensive Care, Rangueil Hospital-University Hospital of Toulouse, 1 Avenue du Professeur Jean Poulhès TSA 50032, 31059, Toulouse Cedex 9, France.,Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France
| |
Collapse
|
23
|
Ding C, Yang J, Van't Veer C, van der Poll T. Bradykinin receptor deficiency or antagonism do not impact the host response during gram-negative pneumonia-derived sepsis. Intensive Care Med Exp 2019; 7:14. [PMID: 30874974 PMCID: PMC6419653 DOI: 10.1186/s40635-019-0228-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 02/27/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Kinins are short peptides with a wide range of proinflammatory properties that are generated from kininogens in the so-called kallikrein-kinin system. Kinins exert their biological activities through stimulation of two distinct receptor subtypes, the kinin or bradykinin B1 and B2 receptors (B1R, B2R). Acute challenge models have implicated B1R and B2R in the pathogenesis of sepsis. However, their role in the host response during sepsis originating from the lung is not known. RESULTS To determine the role of B1R and B2R in pneumonia-derived sepsis, B1R/B2R-deficient mice and wild-type mice treated with the B1R antagonist R-715 or the B2R antagonist HOE-140 were studied after infection with the common gram-negative pathogen Klebsiella pneumoniae via the airways. Neither B1R/B2R deficiency nor B1R or B2R inhibition influenced bacterial growth at the primary site of infection or dissemination to distant body sites. In addition, B1R/B2R deficiency or inhibition did not impact local or systemic inflammatory responses during Klebsiella induced pneumosepsis. CONCLUSIONS These data argue against an important role for kinins in the host response to pneumonia-derived sepsis caused by a clinically relevant pathogen.
Collapse
Affiliation(s)
- Chao Ding
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Room G2-130, 1105 AZ, Amsterdam, the Netherlands
| | - Jack Yang
- Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Room G2-130, 1105 AZ, Amsterdam, the Netherlands
| | - Cornelis Van't Veer
- Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Room G2-130, 1105 AZ, Amsterdam, the Netherlands
| | - Tom van der Poll
- Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Room G2-130, 1105 AZ, Amsterdam, the Netherlands. .,Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
24
|
Ambade AS, Jung B, Lee D, Doods H, Wu D. Triple-tyrosine kinase inhibition attenuates pulmonary arterial hypertension and neointimal formation. Transl Res 2019; 203:15-30. [PMID: 30142307 DOI: 10.1016/j.trsl.2018.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/21/2018] [Accepted: 07/23/2018] [Indexed: 12/29/2022]
Abstract
The present study examined the effects of simultaneous inhibition of vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF) and fibroblast growth factor (FGF) receptor signaling with BIBF1000, a novel triple tyrosine kinase inhibitor on preventing and reversing the progression of severe pulmonary arterial hypertension (PAH) in an experimental model in rats. Left pneumonectomized male Wistar rats were injected with monocrotaline to induce PAH. Treatment with BIBF1000 from day 1 to day 21 after monocrotaline injection attenuated PAH development, as evidenced by lower values for pulmonary artery pressure (mPAP), right ventricular pressure (RVSP), pulmonary arterial neointimal formation, and the ratio of right ventricular weight to left ventricular and septum weight [RV/(LV+S)] on day 21 compared to control rats. Treatment with BIBF1000 from day 21 to day 42 after monocrotaline injection reversed established PAH as shown by normalized values for mPAP and RVSP, RV/(LV+S) ratio, pulmonary arterial occlusion scores, levels of heart and lung fibrosis, as well as improved survival. Treatment with BIBF1000 reduced inflammatory cell recruitment in bronchoalveolar lavage and lung tissues, reduced CD-68 positive macrophages and expression of proliferating cell nuclear antigen in the perivascular areas, and reduced TNF-α and growth factor productions, and inhibited the phosphorylation of AKT and GSK3β in lungs. In addition, BIBF1000 inhibited pulmonary artery smooth muscle cells migration and proliferation from rat pulmonary artery explant cultures. Simultaneous inhibition of VEGF, PDGF, and FGF receptor signaling by BIBF1000 prevents and reverses the progression of severe pulmonary arterial hypertension and vascular remodeling in this experimental model.
Collapse
Affiliation(s)
- Anjira S Ambade
- Department of BIN Convergence Technology, Chonbuk National University, Jeonju, South Korea
| | - Birgit Jung
- Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Dongwon Lee
- Department of BIN Convergence Technology, Chonbuk National University, Jeonju, South Korea
| | - Henri Doods
- Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Dongmei Wu
- Department of BIN Convergence Technology, Chonbuk National University, Jeonju, South Korea; Department of Research, Mount Sinai Medical Center, Miami Beach, Florida.
| |
Collapse
|
25
|
Berkestedt I, Andersson P, Herwald H, Valik JK, Sörensen O, Bodelsson M. Early depletion of contact system in patients with sepsis: a prospective matched control observational study. APMIS 2018; 126:892-898. [PMID: 30397964 DOI: 10.1111/apm.12898] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 09/25/2018] [Indexed: 01/24/2023]
Abstract
Activation of the contact system generates bradykinin from high-molecular-weight kininogen and has been suggested to participate in the pathophysiology of sepsis. To test this, we prospectively measured bradykinin and high-molecular-weight kininogen levels in a cohort of sepsis patients requiring intensive care. From 29 patients meeting criteria for sepsis or septic shock according to Sepsis-3, blood was sampled within 24 h and on the fourth day following admittance to intensive care. Patients planned for neurosurgery served as matched controls. Sequential organ failure assessment score and 90-day mortality was registered. Bradykinin levels (median [interquartile range]) were lower in sepsis patients (79 [62-172] pg/ml) compared to controls (130 [86-255] pg/ml, p < 0.025) and did not correlate with mortality or severity of circulatory derangement. High-molecular-weight kininogen levels were lower in sepsis patients (1.6 [0.8-4.8] densitometry units) compared to controls (4.4 [2.9-7.7] densitometry units, p < 0.001), suggesting previous contact system activation. High-molecular-weight kininogen levels were lower in non-survivors than survivors (p = 0.003) and negatively correlated to severity of circulatory derangement. We conclude that a role for bradykinin in later stages of severe sepsis must be challenged. Low high-molecular-weight kininogen concentrations suggest that the decrease in bradykinin is due to substrate depletion.
Collapse
Affiliation(s)
- Ingrid Berkestedt
- Department of Clinical Sciences Lund, Anaesthesiology and Intensive Care, Lund University, Skane University Hospital, Lund, Sweden
| | - Pia Andersson
- Department ofInfection Medicine, Lund University, Skane University Hospital, Lund, Sweden
| | - Heiko Herwald
- Department ofInfection Medicine, Lund University, Skane University Hospital, Lund, Sweden
| | - John Karlsson Valik
- Infectious Diseases Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Ola Sörensen
- Department of Clinical Sciences Lund, Anaesthesiology and Intensive Care, Lund University, Skane University Hospital, Lund, Sweden
| | - Mikael Bodelsson
- Department of Clinical Sciences Lund, Anaesthesiology and Intensive Care, Lund University, Skane University Hospital, Lund, Sweden
| |
Collapse
|
26
|
Role of Reduced Nitric Oxide in Liver Cell Apoptosis Inhibition During Liver Damage. Arch Med Res 2018; 49:219-225. [DOI: 10.1016/j.arcmed.2018.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/14/2018] [Indexed: 01/05/2023]
|
27
|
Ahmed A, Dolasia K, Mukhopadhyay S. Mycobacterium tuberculosisPPE18 Protein Reduces Inflammation and Increases Survival in Animal Model of Sepsis. THE JOURNAL OF IMMUNOLOGY 2018; 200:3587-3598. [DOI: 10.4049/jimmunol.1602065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/14/2018] [Indexed: 12/16/2022]
|
28
|
Effect of the bradykinin 1 receptor antagonist SSR240612 after oral administration in Mycobacterium tuberculosis-infected mice. Tuberculosis (Edinb) 2018; 109:1-7. [PMID: 29559112 DOI: 10.1016/j.tube.2018.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 01/08/2018] [Accepted: 01/15/2018] [Indexed: 11/21/2022]
Abstract
The role, if any, played by the kinin system in tuberculosis infection models, either in vivo or in vitro, was investigated. The effects of Mycobacterium tuberculosis infection on C57BL/6 wild type, B1R-/-, B2R-/- and double B1R/B2R knockout mice were evaluated. Immunohistochemistry analysis was carried out to assess B1R and B2R expression in spleens and lungs of M. tuberculosis-infected mice. In addition, in vitro experiments with M. tuberculosis-infected macrophages were performed. The in vivo effects of HOE-140 and SSR240612 on the mice model of infection were also evaluated. Infected B2R-/- mice exhibited increased splenomegaly, whereas decreased spleen weight in infected double B1R/B2R knockout mice was observed. The bacterial load, determined as colony-forming units, did not differ in the spleens and lungs of the studied mouse strains. Importantly, immunohistochemical analysis revealed that B1R was upregulated in both spleens and lungs of infected mice. M. tuberculosis-infected macrophages incubated with SSR240612, alone or in combination with des-Arg9-BK, for four days, displayed a marked inhibitory effect on CFU counts. However, the pre-incubation of the selective B1R (des-Arg9-BK and SSR240612) and B2R (BK and HOE-140) agonists and antagonists, respectively, did not significantly affect the bacterial loads. A statistically significant reduction in the CFU of M. tuberculosis in lungs and spleens of animals treated with SSR240612, but not with HOE-140, was observed. Further efforts should be pursued to clarify whether or not SSR240612 might be considered an option for the treatment of tuberculosis.
Collapse
|
29
|
Abstract
INTRODUCTION Kinins are peptide mediators exerting their pro-inflammatory actions by the selective stimulation of two distinct G-protein coupled receptors, termed BKB1R and BKB2R. While BKB2R is constitutively expressed in a multitude of tissues, BKB1R is hardly expressed at baseline but highly inducible by inflammatory mediators. In particular, BKB1R was shown to be involved in the pathogenesis of numerous inflammatory diseases. Areas covered: This review intends to evaluate the therapeutic potential of substances interacting with the BKB1R. To this purpose we summarize the published literature on animal studies with antagonists and knockout mice for this receptor. Expert Opinion: In most cases the pharmacological inhibition of BKB1R or its genetic deletion was beneficial for the outcome of the disease in animal models. Therefore, several companies have developed BKB1R antagonists and tested them in phase I and II clinical trials. However, none of the developed BKB1R antagonists was further developed for clinical use. We discuss possible reasons for this failure of translation of preclinical findings on BKB1R antagonists into the clinic.
Collapse
Affiliation(s)
- Fatimunnisa Qadri
- a Max-Delbrück Center for Molecular Medicine (MDC) , Berlin , Germany
| | - Michael Bader
- a Max-Delbrück Center for Molecular Medicine (MDC) , Berlin , Germany.,b Berlin Institute of Health (BIH) , Berlin , Germany.,c Charité University Medicine Berlin , Germany.,d German Center for Cardiovascular Research (DZHK) site Berlin , Berlin , Germany.,e Institute for Biology , University of Lübeck , Lübeck , Germany
| |
Collapse
|
30
|
Nicola H. The role of contact system in septic shock: the next target? An overview of the current evidence. J Intensive Care 2017; 5:31. [PMID: 28572980 PMCID: PMC5450093 DOI: 10.1186/s40560-017-0228-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/23/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Septic shock remains challenging to intensive care units worldwide, despite recent documented improvement in mortality over the years. Multiple new therapies have been attempted without success in large clinical trials. Evidence concerning the role of the contact system and bradykinin on septic shock physiological manifestations is shown by this article. OBJECTIVES The objective of the study is to review the current evidence linking contact system activation and septic shock, as well as efficacy of available therapies targeting this pathophysiological pathway and to evaluate the potential of further researching the matter. RESULTS Multiple animal studies are already available and suggestive of a meaningful role of contact system activation on septic shock. However, human trials are still scarce, and the ones available are not enough to establish such a strong connection. Furthermore, attempted therapies have been successful across multiple species, but not as much in humans. Therefore, contact system and septic shock relationship remains plentiful in questions to be answered in the coming years or decades. CONCLUSIONS Whether the contact system is not as relevant in humans as it is in animals or there is only lack of evidence remains to be explained. The subject is an attractive open field for further research aiming to aid in tackling such a burdensome condition.
Collapse
Affiliation(s)
- Henrique Nicola
- Intensive Care Registrar Trainee, Royal Perth Hospital, 197 Wellington St Perth, Western Australia, 6000 Australia
| |
Collapse
|