1
|
Kasner SE, Bath PM, Hill MD, Volpi JJ, Giuffre M, Masuoka L, Wambeke D, Madeddu PR. Recombinant Human Tissue Kallikrein-1 for Treating Acute Ischemic Stroke and Preventing Recurrence. Stroke 2025; 56:745-753. [PMID: 39758014 PMCID: PMC11850014 DOI: 10.1161/strokeaha.124.048858] [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] [Indexed: 01/07/2025]
Abstract
Novel strategies are needed for the treatment of acute ischemic stroke when revascularization therapies are not clinically appropriate or are unsuccessful. rKLK1 (recombinant human tissue kallikrein-1), a bradykinin-producing enzyme, offers a promising potential solution. In animal studies of acute stroke, there is a marked 36-fold increase in bradykinin B2 receptor on brain endothelial cells of the ischemic region. Due to this environment, rKLK1-generated bradykinin will exert a potent local vasodilation and increase brain perfusion via 3 synergistic signaling pathways downstream to the B2 receptor. Because of its preferential effect on ischemic tissue, systemic adverse effects such as hypotension are avoided with proper dosing. In addition, with initial vasodilation through recruitment of preexisting collaterals, rKLK1 promotes long-term benefit of brain perfusion by promoting new collateral formation. With an extended course of therapy for weeks after acute ischemic stroke, these multifaceted effects may also reduce the risk of stroke recurrence. A prior phase II trial demonstrated a favorable impact on clinical outcomes and recurrent strokes, particularly among patients who were not eligible for mechanical thrombectomy. A phase II/III trial has launched in this population, though opportunities for combination revascularization therapies deserve further investigation.
Collapse
Affiliation(s)
- Scott E. Kasner
- University of Pennsylvania School of Medicine, Division of Vascular Neurology, Philadelphia (S.E.K.)
| | - Philip M. Bath
- Stroke Trials Unit, University of Nottingham, United Kingdom (P.M.B.)
| | - Michael D. Hill
- Department of Clinical Neuroscience and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary and Foothills Medical Centre, Alberta, Canada (M.D.H.)
| | - John J. Volpi
- Houston Methodist, Department of Neurology, Houston, TX (J.J.V.)
| | - Michael Giuffre
- Faculty of Medicine, University of Calgary, Alberta, Canada (M.G.)
| | | | | | - Paolo R. Madeddu
- Experimental Cardiovascular Medicine, University of Bristol, United Kingdom (P.R.M.)
| |
Collapse
|
2
|
Ji M, Ran X, Zuo H, Zhang Q. Novel Insights into the Kallikrein-Kinin System in Fulminant Myocarditis: Physiological Basis and Potential Therapeutic Advances. J Inflamm Res 2024; 17:7347-7360. [PMID: 39429854 PMCID: PMC11490248 DOI: 10.2147/jir.s488237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024] Open
Abstract
Fulminant myocarditis (FM) is characterized by rapid cardiac deterioration often instigated by an inflammatory cytokine storm. The kallikrein-kinin system (KKS) is a metabolic cascade known for releasing vasoactive kinins, such as bradykinin-related peptides, possessing diverse pharmacological activities that include inflammation, regulation of vascular permeability, endothelial barrier dysfunction, and blood pressure modulation. The type 1 and type 2 bradykinin receptors (B1R and B2R), integral components of the KKS system, mediate the primary biological effects of kinin peptides. This review aims to offer a comprehensive overview of the primary mechanisms of the KKS in FM, including an examination of the structural components, regulatory activation, and downstream signaling pathways of the KKS. Furthermore, it explores the involvement of the tissue kallikrein/B1R/inducible nitric oxide synthase (TK/B1R/iNOS) pathway in myocyte dysfunction, modulation of the immune response, and preservation of endothelial barrier integrity. The potential therapeutic advances targeting the inhibition of the KKS in managing FM will be discussed, providing valuable insights for the development of clinical treatment strategies.
Collapse
Affiliation(s)
- Mengmeng Ji
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Xiao Ran
- Department of Critical-Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Houjuan Zuo
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Qin Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
3
|
Oliveira AC, Vicentino ARR, Andrade D, Pereira IR, Saboia-Vahia L, Moreira ODC, Carvalho-Pinto CE, Mota JBD, Maciel L, Vilar-Pereira G, Pesquero JB, Lannes-Vieira J, Sirois P, Campos de Carvalho AC, Scharfstein J. Genetic Ablation and Pharmacological Blockade of Bradykinin B1 Receptor Unveiled a Detrimental Role for the Kinin System in Chagas Disease Cardiomyopathy. J Clin Med 2023; 12:jcm12082888. [PMID: 37109224 PMCID: PMC10144326 DOI: 10.3390/jcm12082888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/24/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Chagas disease, the parasitic infection caused by Trypanosoma cruzi, afflicts about 6 million people in Latin America. Here, we investigated the hypothesis that T. cruzi may fuel heart parasitism by activating B1R, a G protein-coupled (brady) kinin receptor whose expression is upregulated in inflamed tissues. Studies in WT and B1R-/- mice showed that T. cruzi DNA levels (15 days post infection-dpi) were sharply reduced in the transgenic heart. FACS analysis revealed that frequencies of proinflammatory neutrophils and monocytes were diminished in B1R-/- hearts whereas CK-MB activity (60 dpi) was exclusively detected in B1R+/+ sera. Since chronic myocarditis and heart fibrosis (90 dpi) were markedly attenuated in the transgenic mice, we sought to determine whether a pharmacological blockade of the des-Arg9-bradykinin (DABK)/B1R pathway might alleviate chagasic cardiomyopathy. Using C57BL/6 mice acutely infected by a myotropic T. cruzi strain (Colombian), we found that daily treatment (15-60 dpi) with R-954 (B1R antagonist) reduced heart parasitism and blunted cardiac injury. Extending R-954 treatment to the chronic phase (120-160 dpi), we verified that B1R targeting (i) decreased mortality indexes, (ii) mitigated chronic myocarditis, and (iii) ameliorated heart conduction disturbances. Collectively, our study suggests that a pharmacological blockade of the proinflammatory KKS/DABK/B1R pathway is cardioprotective in acute and chronic Chagas disease.
Collapse
Affiliation(s)
- Ana Carolina Oliveira
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Amanda Roberta Revoredo Vicentino
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Daniele Andrade
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Isabela Resende Pereira
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
| | - Leonardo Saboia-Vahia
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Otacílio da Cruz Moreira
- Plataforma de PCR em Tempo Real RPT09A, Laboratório de Virologia Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
| | - Carla Eponina Carvalho-Pinto
- Departamento de Imunobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói 24020-141, Brazil
| | - Julia Barbalho da Mota
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Leonardo Maciel
- Programa de Medicina Regenerativa, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Núcleo Multidisciplinar de Pesquisa em Biologia, Universidade Federal do Rio de Janeiro, Duque de Caxias Campus, Rio de Janeiro 21941-902, Brazil
| | - Glaucia Vilar-Pereira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
| | - João B Pesquero
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo 05508-090, Brazil
| | - Joseli Lannes-Vieira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
| | - Pierre Sirois
- Department of Microbiology and Immunology, Faculty of Medicine, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Antônio Carlos Campos de Carvalho
- Programa de Medicina Regenerativa, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Centro Nacional de Biologia Estrutural e Bio-Imagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro 21941-902, Brazil
| | - Julio Scharfstein
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| |
Collapse
|
4
|
Othman R, Cagnone G, Joyal JS, Vaucher E, Couture R. Kinins and Their Receptors as Potential Therapeutic Targets in Retinal Pathologies. Cells 2021; 10:1913. [PMID: 34440682 PMCID: PMC8391508 DOI: 10.3390/cells10081913] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 12/29/2022] Open
Abstract
The kallikrein-kinin system (KKS) contributes to retinal inflammation and neovascularization, notably in diabetic retinopathy (DR) and neovascular age-related macular degeneration (AMD). Bradykinin type 1 (B1R) and type 2 (B2R) receptors are G-protein-coupled receptors that sense and mediate the effects of kinins. While B2R is constitutively expressed and regulates a plethora of physiological processes, B1R is almost undetectable under physiological conditions and contributes to pathological inflammation. Several KKS components (kininogens, tissue and plasma kallikreins, and kinin receptors) are overexpressed in human and animal models of retinal diseases, and their inhibition, particularly B1R, reduces inflammation and pathological neovascularization. In this review, we provide an overview of the KKS with emphasis on kinin receptors in the healthy retina and their detrimental roles in DR and AMD. We highlight the crosstalk between the KKS and the renin-angiotensin system (RAS), which is known to be detrimental in ocular pathologies. Targeting the KKS, particularly the B1R, is a promising therapy in retinal diseases, and B1R may represent an effector of the detrimental effects of RAS (Ang II-AT1R).
Collapse
Affiliation(s)
- Rahmeh Othman
- School of Optometry, Université de Montréal, Montreal, QC H3T 1P1, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Gael Cagnone
- Department of Pediatry, Faculty of Medicine, CHU St Justine, Université de Montréal, Montreal, QC H3T 1J4, Canada; (G.C.); (J.-S.J.)
| | - Jean-Sébastien Joyal
- Department of Pediatry, Faculty of Medicine, CHU St Justine, Université de Montréal, Montreal, QC H3T 1J4, Canada; (G.C.); (J.-S.J.)
| | - Elvire Vaucher
- School of Optometry, Université de Montréal, Montreal, QC H3T 1P1, Canada
| | - Réjean Couture
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
5
|
Shen M, Yu M, Qiu C, Zhang G, Li J, Fang W, Wang Q. Myocardial angiogenesis induced by exercise training involves a regulatory mechanism mediated by kinin receptors. Clin Exp Hypertens 2021; 43:408-415. [PMID: 33687297 DOI: 10.1080/10641963.2021.1896725] [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: 11/25/2020] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To demonstrate that the kallikrein-kinin system (KKS) is upstream of angiogenic signaling pathway, and to determine the role of the kinin B1 and B2 receptors in myocardial angiogenesis induced by exercise training. METHODS Forty Wistar rats were randomly assigned to an exercise control (EC) group, a B1 receptor antagonist (B1Ant) group, a B2 receptor antagonist (B2Ant) group, and a double receptor antagonist ((B1+ B2)Ant) group. A myocardial infarction model was employed. Animals in all groups received 30 min of exercise training for 4 weeks. The expression of VEGF and eNOS, capillary supply, and apoptosis rate were evaluated. RESULTS The mRNA and protein expression of VEGF and eNOS showed similar trends in all groups, and were lowest in the (B1+ B2) Ant group, and highest in the EC group. Levels of VEGF and eNOS mRNA were significantly lower in the B1Ant group than in the B2Ant group (p< .001 and p< .05, respectively). VEGF and eNOS protein in the B1Ant group was also significantly lower (p< .01 and p< .05, respectively) than in the B2Ant group. The capillary numbers in the (B1+ B2) Ant group were significantly lower than in the EC group (395.8 ± 105 vs. 1127.9 ± 192.98, respectively). The apoptosis rate of cardiomyocytes was highest in the (B1+ B2) Ant group. CONCLUSION KKS may act as an upstream signal transduction pathway for angiogenic factors in myocardial angiogenesis. The B1 and B2 receptors exert additive effects, and the B1 receptor has the most prominent role in mediating KKS-induced myocardial angiogenesis.
Collapse
MESH Headings
- Animals
- Capillaries/metabolism
- Kinins/metabolism
- Male
- Myocardium/metabolism
- Myocytes, Cardiac/metabolism
- Neovascularization, Physiologic
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- Physical Conditioning, Animal
- Platelet Endothelial Cell Adhesion Molecule-1/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Wistar
- Receptor, Bradykinin B1/genetics
- Receptor, Bradykinin B1/metabolism
- Receptor, Bradykinin B2/genetics
- Receptor, Bradykinin B2/metabolism
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
- Rats
Collapse
Affiliation(s)
- Mei Shen
- Department of Rehabilitation Medicin, The People's Hospital of Longhua District, Shenzhen, Guangdong Province, China
| | - Min Yu
- Department of Rehabilitation Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning Province, China
| | - Chengxiu Qiu
- Department of Rehabilitation Medicin, The People's Hospital of Longhua District, Shenzhen, Guangdong Province, China
| | - Ge Zhang
- Department of Electrocardiogram, The People's Hospital of Longhua District, Shenzhen, Guangdong Province, China
| | - Jingya Li
- Department of Rehabilitation Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning Province, China
| | - Wei Fang
- Department of Nursing, The People's Hospital of Longhua District, Shenzhen, Guangdong Province, China
| | - Qiwen Wang
- Department of Rehabilitation Medicin, The People's Hospital of Longhua District, Shenzhen, Guangdong Province, China
| |
Collapse
|
6
|
Differential Expression of Kinin Receptors in Human Wet and Dry Age-Related Macular Degeneration Retinae. Pharmaceuticals (Basel) 2020; 13:ph13060130. [PMID: 32599742 PMCID: PMC7345220 DOI: 10.3390/ph13060130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/08/2020] [Accepted: 06/22/2020] [Indexed: 11/17/2022] Open
Abstract
Kinins are vasoactive peptides and mediators of inflammation, which signal through two G protein-coupled receptors, B1 and B2 receptors (B1R, B2R). Recent pre-clinical findings suggest a primary role for B1R in a rat model of wet age-related macular degeneration (AMD). The aim of the present study was to investigate whether kinin receptors are differentially expressed in human wet and dry AMD retinae. The cellular distribution of B1R and B2R was examined by immunofluorescence and in situ hybridization in post-mortem human AMD retinae. The association of B1R with inflammatory proteins (inducible nitric oxide synthase (iNOS) and vascular endothelial growth factor A (VEGFA)), fibrosis markers and glial cells was also studied. While B2R mRNA and protein expression was not affected by AMD, a significant increase of B1R mRNA and immunoreactivity was measured in wet AMD retinae when compared to control and dry AMD retinae. B1R was expressed by Müller cells, astrocytes, microglia and endothelial/vascular smooth muscle cells, and colocalized with iNOS and fibrosis markers, but not with VEGFA. In conclusion, the induction and upregulation of the pro-inflammatory and pro-fibrotic kinin B1R in human wet AMD retinae support previous pre-clinical studies and provide a clinical proof-of-concept that B1R represents an attractive therapeutic target worth exploring in this retinal disease.
Collapse
|
7
|
Hillmeister P, Bondke Persson A. Bradykinin-from snake poison to therapeutic options. Acta Physiol (Oxf) 2020; 228:e13445. [PMID: 31950593 DOI: 10.1111/apha.13445] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 01/11/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Philipp Hillmeister
- Campus Clinic Brandenburg, Internal Medicine 1, Department for Angiology, Faculty of Health Sciences joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg the Brandenburg Medical School Theodor Fontane and the University of Potsdam Brandenburg an der Havel Germany
| | - Anja Bondke Persson
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health, Institute of Vegetative Physiology Berlin Germany
| |
Collapse
|
8
|
Hachana S, Fontaine O, Sapieha P, Lesk M, Couture R, Vaucher E. The effects of anti-VEGF and kinin B 1 receptor blockade on retinal inflammation in laser-induced choroidal neovascularization. Br J Pharmacol 2020; 177:1949-1966. [PMID: 31883121 DOI: 10.1111/bph.14962] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Age-related macular degeneration (AMD) is a complex neurodegenerative disease treated by anti-VEGF intravitreal injections. As inflammation is potentially involved in retinal degeneration, the pro-inflammatory kallikrein-kinin system is a possible alternative pharmacological target. Here, we investigated the effects of anti-VEGF and anti-B1 receptor treatments on the inflammatory mechanisms in a rat model of choroidal neovascularization (CNV). EXPERIMENTAL APPROACH Immediately after laser-induced CNV, Long-Evans rats were treated by eye-drop application of a B1 receptor antagonist (R-954) or by intravitreal injection of B1 receptor siRNA or anti-VEGF antibodies. Effects of treatments on gene expression of inflammatory mediators, CNV lesion regression and integrity of the blood-retinal barrier was measured 10 days later in the retina. B1 receptor and VEGF-R2 cellular localization was assessed. KEY RESULTS The three treatments significantly inhibited the CNV-induced retinal changes. Anti-VEGF and R-954 decreased CNV-induced up-regulation of B1 and B2 receptors, TNF-α, and ICAM-1. Anti-VEGF additionally reversed up-regulation of VEGF-A, VEGF-R2, HIF-1α, CCL2 and VCAM-1, whereas R-954 inhibited gene expression of IL-1β and COX-2. Enhanced retinal vascular permeability was abolished by anti-VEGF and reduced by R-954 and B1 receptor siRNA treatments. Leukocyte adhesion was impaired by anti-VEGF and B1 receptor inhibition. B1 receptors were found on astrocytes and endothelial cells. CONCLUSION AND IMPLICATIONS B1 receptor and VEGF pathways were both involved in retinal inflammation and damage in laser-induced CNV. The non-invasive, self-administration of B1 receptor antagonists on the surface of the cornea by eye drops might be an important asset for the treatment of AMD.
Collapse
Affiliation(s)
- Soumaya Hachana
- School of Optometry, Université de Montréal, Montréal, Quebec, Canada.,Department of Pharmacology and Physiology, Université de Montréal, Montréal, Quebec, Canada
| | - Olivier Fontaine
- School of Optometry, Université de Montréal, Montréal, Quebec, Canada.,Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Montréal, Quebec, Canada
| | - Przemyslaw Sapieha
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Montréal, Quebec, Canada
| | - Mark Lesk
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Montréal, Quebec, Canada
| | - Réjean Couture
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Quebec, Canada
| | - Elvire Vaucher
- School of Optometry, Université de Montréal, Montréal, Quebec, Canada
| |
Collapse
|
9
|
Bhatwadekar AD, Kansara VS, Ciulla TA. Investigational plasma kallikrein inhibitors for the treatment of diabetic macular edema: an expert assessment. Expert Opin Investig Drugs 2020; 29:237-244. [PMID: 31985300 DOI: 10.1080/13543784.2020.1723078] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Plasma kallikrein is a mediator of vascular leakage and inflammation. Activation of plasma kallikrein can induce features of diabetic macular edema (DME) in preclinical models. Human vitreous shows elevated plasma kallikrein levels in patients with DME. Because of the incomplete response of some patients to anti-VEGF agents, and the treatment burden associated with frequent dosing, there is still considerable need for VEGF-independent targeted pathways.Areas covered: This review covers the role of plasma kallikrein in the pathogenesis of DME and the therapeutic potential of plasma kallikrein inhibitors. It discusses early clinical studies of plasma kallikrein pathway modulation for DME, which have been associated with some improvement in visual acuity but with limited improvement in macular edema. This review also highlights KVD001, which is furthest along the development pathway, THR-149, which has recently completed a phase 1 study, and oral agents under development.Expert opinion: Plasma kallikrein inhibitors have a potential role in the treatment of DME, with mixed functional/anatomic results in early clinical trials. Given the large unmet need in DME treatment, further studies are warranted.
Collapse
Affiliation(s)
- Ashay D Bhatwadekar
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, IN, USA
| | | | - Thomas A Ciulla
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, IN, USA.,Clearside Biomedical, Inc., Alpharetta, GA, USA.,Retina Service, Midwest Eye Institute, Indianapolis, IN, USA
| |
Collapse
|
10
|
Maria AG, Dillemburg-Pilla P, Durand MDT, Floriano EM, Manfiolli AO, Ramos SG, Pesquero JB, Nahmias C, Costa-Neto CM. Activation of the Kinin B1 Receptor by Its Agonist Reduces Melanoma Metastasis by Playing a Dual Effect on Tumor Cells and Host Immune Response. Front Pharmacol 2019; 10:1106. [PMID: 31607931 PMCID: PMC6774293 DOI: 10.3389/fphar.2019.01106] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 08/28/2019] [Indexed: 12/20/2022] Open
Abstract
Metastatic melanoma is an aggressive type of skin cancer leading half of the patients to death within 8–10 months after diagnosis. Kinins are peptides that interact with B1 and B2 receptors playing diverse biological roles. We investigated whether treatment with B1 receptor agonist, des-Arg9-bradykinin (DABK), has effects in lung metastasis establishment after melanoma induction in mice. We found a lower number of metastatic colonies in lungs of DABK-treated mice, reduced expression of vascular cell adhesion molecule 1 (VCAM-1), and increased CD8+T-cell recruitment to the metastatic area compared to animals that did not receive treatment. To understand whether the effects of DABK observed were due to the activation of the B1 receptor in the tumor cells or in the host, we treated wild-type (WT) and kinin B1 receptor knockout (B1−/−) mice with DABK. No significant differences in the number of melanoma colonies established in lungs were seen between WT and B1−/−mice; however, B1−/−mice presented higher VCAM-1 expression and lower CD8+T-cell infiltration. In conclusion, we believe that activation of kinin B1 receptor by its agonist in the host stimulates the immune response more efficiently, promoting CD8+T-cell recruitment to the metastatic lungs and interfering in VCAM-1 expression. Moreover, treatment with DABK reduced establishment of metastatic colonies by mainly acting on tumor cells; hence, this study brings insights to explore novel approaches to treat metastatic melanoma targeting the B1 receptor.
Collapse
Affiliation(s)
- Andrea Gutierrez Maria
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Patrícia Dillemburg-Pilla
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Elaine Medeiros Floriano
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Adriana Oliveira Manfiolli
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Simone Gusmão Ramos
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João Bosco Pesquero
- Department of Biophysics, Federal University of São Paulo, São Paulo, Brazil
| | - Clara Nahmias
- INSERM U981, Department of Molecular Medicine, Gustave Roussy Cancer Center, Villejuif, France
| | - Claudio M Costa-Neto
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
11
|
Shen M, Yu M, Li J, Ma L. Effects of exercise training on kinin receptors expression in rats with myocardial infarction. Arch Physiol Biochem 2017; 123:206-211. [PMID: 28330378 DOI: 10.1080/13813455.2017.1302962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVES The objective of this study is to determine the role of kinin B1 and B2 receptors in exercise-induced cardiac muscle angiogenesis. METHOD Thirty Wistar rats were randomly assigned to the control group, the myocardial infarction group and the exercise training group (myocardial infarction model was made and received 30 min exercise training on a treadmill). After 4 weeks of experiment, cardiac muscle was harvested. RESULTS B1 and B2 receptor mRNA and protein levels in the exercise-training group were significantly higher than those in the myocardial infarction group, which were higher than those in the control group. Capillary number in the cardiac muscle also showed the same tendency. There was a correlation between capillary number and B1 receptor protein (not B2 receptor protein) in the all groups. CONCLUSION Kinin B1 and B2 receptors play roles in exercise-induced cardiac muscle angiogenesis. However, the B1 receptor appears to have a more prominent role.
Collapse
Affiliation(s)
- Mei Shen
- a Department of Rehabilitation Medicine , Affiliated Zhongshan Hospital of Dalian University , Dalian , China
| | - Min Yu
- a Department of Rehabilitation Medicine , Affiliated Zhongshan Hospital of Dalian University , Dalian , China
| | - Jingya Li
- a Department of Rehabilitation Medicine , Affiliated Zhongshan Hospital of Dalian University , Dalian , China
| | - Li Ma
- a Department of Rehabilitation Medicine , Affiliated Zhongshan Hospital of Dalian University , Dalian , China
| |
Collapse
|
12
|
Intracellular signaling pathways involved in the release of IL-4 and VEGF from human keratinocytes by activation of kinin B1 receptor: functional relevance to angiogenesis. Arch Dermatol Res 2015; 307:803-17. [PMID: 26338700 DOI: 10.1007/s00403-015-1595-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 08/04/2015] [Accepted: 08/20/2015] [Indexed: 12/11/2022]
Abstract
The injured skin produces a number of mediators that directly or indirectly modulate cell chemotaxis, migration, proliferation, and angiogenesis. Components of the kinin pathway including the kinin B1 receptor (B1R) have been found to occur in the human skin, but information about its role on keratinocyte biology is still scarce. Our aim was to determine whether stimulation of B1R causes the secretion of IL-4 and/or VEGF from human keratinocytes and to evaluate the role of the B1R agonist Lys-des[Arg(9)]bradykinin and IL-4 on various stages of angiogenesis, such as cell migration, proliferation, and release of metalloproteases. By using ELISA and Western blotting, we showed that HaCaT keratinocytes stimulated with the B1R agonist release IL-4 and VEGF. Stimulation of B1R also caused transient c-JunN-terminal kinase phosphorylation and JunB nuclear translocation, transcription factor that regulates IL-4 expression. The 3D-angiogenesis assay, performed on spheroids of EA.hy923 endothelial cells embedded in a collagen matrix, showed that their cumulative sprout area increased significantly following stimulation with either IL-4 or B1R agonist. Furthermore, these ligands produced significant endothelial cell migration and release of metalloproteases 2 and 9, but did not increase endothelial cell proliferation as measured by 5-bromo-2'-deoxyuridine incorporation. Our results provide experimental evidence that establishes IL-4 and B1R agonist as important angiogenic factors of relevance for skin repair.
Collapse
|
13
|
Desposito D, Potier L, Chollet C, Gobeil F, Roussel R, Alhenc-Gelas F, Bouby N, Waeckel L. Kinin receptor agonism restores hindlimb postischemic neovascularization capacity in diabetic mice. J Pharmacol Exp Ther 2015; 352:218-26. [PMID: 25398240 DOI: 10.1124/jpet.114.219196] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Limb ischemia is a major complication of thromboembolic diseases. Diabetes worsens prognosis by impairing neovascularization. Genetic or pharmacological inactivation of the kallikrein-kinin system aggravates limb ischemia in nondiabetic animals, whereas angiotensin I-converting enzyme/kininase II inhibition improves outcome. The role of kinins in limb ischemia in the setting of diabetes is not documented. We assessed whether selective activation of kinin receptors by pharmacological agonists can influence neovascularization in diabetic mice with limb ischemia and have a therapeutic effect. Selective pseudopeptide kinin B1 or B2 receptor agonists resistant to peptidase action were administered by osmotic minipumps at a nonhypotensive dosage for 14 days after unilateral femoral artery ligation in mice previously rendered diabetic by streptozotocin. Comparison was made with ligatured, nonagonist-treated nondiabetic and diabetic mice. Diabetes reduced neovascularization, assessed by microangiography and histologic capillary density analysis, by roughly 40%. B1 receptor agonist or B2 receptor agonist similarly restored neovascularization in diabetic mice. Neovascularization in agonist-treated diabetic mice was indistinguishable from nondiabetic mice. Both treatments restored blood flow in the ischemic hindfoot, measured by laser-Doppler perfusion imaging. Macrophage infiltration increased 3-fold in the ischemic gastrocnemius muscle during B1 receptor agonist or B2 receptor agonist treatment, and vascular endothelial growth factor (VEGF) level increased 2-fold. Both treatments increased, by 50-100%, circulating CD45/CD11b-positive monocytes and CD34(+)/VEGFR2(+) progenitor cells. Thus, selective pharmacological activation of B1 or B2 kinin receptor overcomes the effect of diabetes on postischemic neovascularization and restores tissue perfusion through monocyte/macrophage mobilization. Kinin receptors are potential therapeutic targets in limb ischemia in diabetes.
Collapse
Affiliation(s)
- Dorinne Desposito
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Louis Potier
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Catherine Chollet
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Fernand Gobeil
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Ronan Roussel
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Francois Alhenc-Gelas
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Nadine Bouby
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Ludovic Waeckel
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| |
Collapse
|
14
|
Naidu N, Botha JH, Naidoo S. B1 but not B2 bradykinin receptor agonists promote DU145 prostate cancer cell proliferation and migration. Afr Health Sci 2014; 14:657-62. [PMID: 25352885 DOI: 10.4314/ahs.v14i3.22] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The kallikrein-kinin system (KKS) is an endogenous pathway involved in angiogenesis and tumourigenesis, both vital for cancer growth and progression. OBJECTIVES To investigate the effect of two bradykinin receptor (B1R and B2R) agonists on growth and motility of prostate tumour (DU145) and micro-vascular endothelial cells (dMVECs). METHODS Increasing concentrations of selective B1R and B2R agonists were added to cultured cells. Cell proliferation and migration were assessed using the 3-[4,5 dimethylthiazol-2-yl]-2,5-diphenyl-tetrazolium bromide (MTT) and modified Boyden Chamber assays, respectively. Where significant stimulation was found, the influence of an antagonist was also investigated. RESULTS Neither growth nor motility of endothelial cells was affected by either agonist. In DU145 cells, while the B2R agonist was without any significant effect, the B1R agonist stimulated proliferation and migration at concentrations of 10nM and 50nM respectively. Further, this effect was abrogated when cells were pre-incubated with a B1R antagonist. CONCLUSIONS Unlike the physiologically-active B2R, the pathologically-inducible B1R may be implicated in prostate tumourigenic events. The involvement of the KKS in malignant prostate pathology supports on-going exploration of bradykinin receptor antagonists as target candidates in the development of alternate approaches to cancer therapy.
Collapse
Affiliation(s)
- N Naidu
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - J H Botha
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - S Naidoo
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| |
Collapse
|
15
|
Marketou ME, Kontaraki J, Zacharis E, Parthenakis F, Maragkoudakis S, Gavras I, Gavras H, Vardas PE. Differential gene expression of bradykinin receptors 1 and 2 in peripheral monocytes from patients with essential hypertension. J Hum Hypertens 2014; 28:450-5. [PMID: 24401952 DOI: 10.1038/jhh.2013.133] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 11/07/2013] [Accepted: 11/19/2013] [Indexed: 12/26/2022]
Abstract
Bradykinin participates in various hypertensive processes, exerted via its type 1 and type 2 receptors (BKR1 and BKR2). The aim of the study was to investigate BKR1 and BK2R gene expression in peripheral monocytes in patients with essential hypertension compared with healthy individuals. Seventeen hypertensive patients (9 males, age 56 ± 7 years) and 12 healthy individuals (7 males, age 55 ± 6) participated. Mononuclear cells isolated using anti-CD14+ antibodies and mRNAs of BKR1 and BKR2 were estimated by real-time quantitative reverse transcription-PCR. Both BKR1 and BKR2 showed significantly upregulated gene expression in the group of hypertensive patients. Specifically, BKR1 gene expression was 142.1 ± 42.2 in hypertensives versus 20.2 ± 8 in controls (P = 0.024) and BKR2 was 1222.2 ± 361.6 in hypertensives versus 259.5 ± 99.1 in controls (P = 0.038). Antihypertensive treatment resulted in a decrease in BKR1 (from 142.1 ± 42.2 to 55.2 ± 17.1, P = 0.065) and in BKR2 (from 1222.2 ± 361.6 to 256.8 ± 81.8, P = 0.014) gene expression. BKR1 and BKR2 gene expression on peripheral monocytes is upregulated in essential hypertension. This may lead to functional changes in monocytes and contribute to the development of target organ damage in hypertensive patients.
Collapse
Affiliation(s)
- M E Marketou
- Cardiology Department, Heraklion University Hospital, Crete, Greece
| | - J Kontaraki
- Cardiology Department, Heraklion University Hospital, Crete, Greece
| | - E Zacharis
- Cardiology Department, Heraklion University Hospital, Crete, Greece
| | - F Parthenakis
- Cardiology Department, Heraklion University Hospital, Crete, Greece
| | - S Maragkoudakis
- Cardiology Department, Heraklion University Hospital, Crete, Greece
| | - I Gavras
- Hypertension and Atherosclerosis Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - H Gavras
- Hypertension and Atherosclerosis Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - P E Vardas
- Cardiology Department, Heraklion University Hospital, Crete, Greece
| |
Collapse
|
16
|
Girolami JP, Blaes N, Bouby N, Alhenc-Gelas F. Genetic manipulation and genetic variation of the kallikrein-kinin system: impact on cardiovascular and renal diseases. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2014; 69:145-196. [PMID: 25130042 DOI: 10.1007/978-3-319-06683-7_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Genetic manipulation of the kallikrein-kinin system (KKS) in mice, with either gain or loss of function, and study of human genetic variability in KKS components which has been well documented at the phenotypic and genomic level, have allowed recognizing the physiological role of KKS in health and in disease. This role has been especially documented in the cardiovascular system and the kidney. Kinins are produced at slow rate in most organs in resting condition and/or inactivated quickly. Yet the KKS is involved in arterial function and in renal tubular function. In several pathological situations, kinin production increases, kinin receptor synthesis is upregulated, and kinins play an important role, whether beneficial or detrimental, in disease outcome. In the setting of ischemic, diabetic or hemodynamic aggression, kinin release by tissue kallikrein protects against organ damage, through B2 and/or B1 bradykinin receptor activation, depending on organ and disease. This has been well documented for the ischemic or diabetic heart, kidney and skeletal muscle, where KKS activity reduces oxidative stress, limits necrosis or fibrosis and promotes angiogenesis. On the other hand, in some pathological situations where plasma prekallikrein is inappropriately activated, excess kinin release in local or systemic circulation is detrimental, through oedema or hypotension. Putative therapeutic application of these clinical and experimental findings through current pharmacological development is discussed in the chapter.
Collapse
|
17
|
Bradykinin in health and disease: proceedings of the Bradykinin Symposium 2012, Berlin 23-24 August 2012. Inflamm Res 2013; 63:173-8. [PMID: 24316865 DOI: 10.1007/s00011-013-0693-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 11/26/2013] [Indexed: 10/25/2022] Open
|
18
|
Zhang X, Tan F, Skidgel RA. Carboxypeptidase M is a positive allosteric modulator of the kinin B1 receptor. J Biol Chem 2013; 288:33226-40. [PMID: 24108126 DOI: 10.1074/jbc.m113.520791] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ligand binding to extracellular domains of G protein-coupled receptors can result in novel and nuanced allosteric effects on receptor signaling. We previously showed that the protein-protein interaction of carboxypeptidase M (CPM) and kinin B1 receptor (B1R) enhances B1R signaling in two ways; 1) kinin binding to CPM causes a conformational activation of the B1R, and 2) CPM-generated des-Arg-kinin agonist is efficiently delivered to the B1R. Here, we show CPM is also a positive allosteric modulator of B1R signaling to its agonist, des-Arg(10)-kallidin (DAKD). In HEK cells stably transfected with B1R, co-expression of CPM enhanced DAKD-stimulated increases in intracellular Ca(2+) or phosphoinositide turnover by a leftward shift of the dose-response curve without changing the maximum. CPM increased B1R affinity for DAKD by ∼5-fold but had no effect on basal B1R-dependent phosphoinositide turnover. Soluble, recombinant CPM bound to HEK cells expressing B1Rs without stimulating receptor signaling. CPM positive allosteric action was independent of enzyme activity but depended on interaction of its C-terminal domain with the B1R extracellular loop 2. Disruption of the CPM/B1R interaction or knockdown of CPM in cytokine-treated primary human endothelial cells inhibited the allosteric enhancement of CPM on B1R DAKD binding or ERK1/2 activation. CPM also enhanced the DAKD-induced B1R conformational change as detected by increased intramolecular fluorescence or bioluminescence resonance energy transfer. Thus, CPM binding to extracellular loop 2 of the B1R results in positive allosteric modulation of B1R signaling, and disruption of this interaction could provide a novel therapeutic approach to reduce pathological B1R signaling.
Collapse
|
19
|
Su JB. Different cross-talk sites between the renin-angiotensin and the kallikrein-kinin systems. J Renin Angiotensin Aldosterone Syst 2013; 15:319-28. [PMID: 23386283 DOI: 10.1177/1470320312474854] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Targeting the renin-angiotensin system (RAS) constitutes a major advance in the treatment of cardiovascular diseases. Evidence indicates that angiotensin-converting enzyme inhibitors and angiotensin AT1 receptor blockers act on both the RAS and the kallikrein-kinin system (KKS). In addition to the interaction between the RAS and KKS at the level of angiotensin-converting enzyme catalyzing both angiotensin II generation and bradykinin degradation, the RAS and KKS also interact at other levels: 1) prolylcarboxypeptidase, an angiotensin II inactivating enzyme and a prekallikrein activator; 2) kallikrein, a kinin-generating and prorenin-activating enzyme; 3) angiotensin-(1-7) exerts kininlike effects and potentiates the effects of bradykinin; and 4) the angiotensin AT1 receptor forms heterodimers with the bradykinin B2 receptor. Moreover, angiotensin II enhances B1 and B2 receptor expression via transcriptional mechanisms. These cross-talks explain why both the RAS and KKS are up-regulated in some circumstances, whereas in other circumstances both systems change in the opposite manner, expressed as an activated RAS and a depressed KKS. As the cross-talks between the RAS and the KKS play an important role in response to different stimuli, taking these cross-talks between the two systems into account may help in the development of drugs targeting the two systems.
Collapse
Affiliation(s)
- Jin Bo Su
- Inserm U955, Maisons-Alfort, France, and Faculté de Médecine de Créteil, Université Paris-Est, France
| |
Collapse
|
20
|
Effects of a novel bradykinin B1 receptor antagonist and angiotensin II receptor blockade on experimental myocardial infarction in rats. PLoS One 2012; 7:e51151. [PMID: 23236443 PMCID: PMC3517424 DOI: 10.1371/journal.pone.0051151] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 10/29/2012] [Indexed: 01/06/2023] Open
Abstract
Background The aim of the present study was to evaluate the cardiovascular effects of the novel bradykinin B1 receptor antagonist BI-113823 following myocardial infarction (MI) and to determine whether B1 receptor blockade alters the cardiovascular effects of an angiotensin II type 1 (AT1) receptor antagonist after MI in rats. Methodology/Principal Findings Sprague Dawley rats were subjected to permanent occlusion of the left descending coronary artery. Cardiovascular function was determined at 7 days post MI. Treatment with either B1 receptor antagonist (BI-113823) or AT1 receptor antagonist (irbesartan) alone or in combination improved post-MI cardiac function as evidenced by attenuation of elevated left ventricular end diastolic pressure (LVEDP); greater first derivative of left ventricular pressure (± dp/dt max), left ventricle ejection fraction, fractional shorting, and better wall motion; as we as reductions in post-MI up-regulation of matrix metalloproteinases 2 (MMP-2) and collagen III. In addition, the cardiac up-regulation of B1 receptor and AT1 receptor mRNA were markedly reduced in animals treated with BI 113823, although bradykinin B2 receptor and angiotensin 1 converting enzyme (ACE1) mRNA expression were not significantly affected by B1 receptor blockade. Conclusions/Significance The present study demonstrates that treatment with the novel B1 receptor antagonist, BI-113823 improves post-MI cardiac function and does not influence the cardiovascular effects of AT1 receptor antagonist following MI.
Collapse
|
21
|
Affiliation(s)
- P. Hillmeister
- Experimental and Clinical Research Center; Center for Cardiovascular Research; Charité-Universitaetsmedizin Berlin; Berlin; Germany
| | - P. B. Persson
- Institute of Vegetative Physiology; Charité-Universitaetsmedizin Berlin; Berlin; Germany
| |
Collapse
|
22
|
Katsi V, Katsimichas T, Pittaras A, Grassos C, Katsimichas A, Tousoulis D, Stefanadis C, Kallikazaros I. Hypertension and bradykinin. Cardiovasc Endocrinol 2012; 1:24-30. [DOI: 10.1097/xce.0b013e328357a94c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
23
|
Maurer M, Bader M, Bas M, Bossi F, Cicardi M, Cugno M, Howarth P, Kaplan A, Kojda G, Leeb-Lundberg F, Lötvall J, Magerl M. New topics in bradykinin research. Allergy 2011; 66:1397-406. [PMID: 21859431 DOI: 10.1111/j.1398-9995.2011.02686.x] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Bradykinin has been implicated to contribute to allergic inflammation and the pathogenesis of allergic conditions. It binds to endothelial B(1) and B(2) receptors and exerts potent pharmacological and physiological effects, notably, decreased blood pressure, increased vascular permeability and the promotion of classical symptoms of inflammation such as vasodilation, hyperthermia, oedema and pain. Towards potential clinical benefit, bradykinin has also been shown to exert potent antithrombogenic, antiproliferative and antifibrogenic effects. The development of pharmacologically active substances, such as bradykinin receptor blockers, opens up new therapeutic options that require further research into bradykinin. This review presents current understanding surrounding the role of bradykinin in nonallergic angioedema and other conditions seen by allergists and emergency physicians, and its potential role as a therapeutic target.
Collapse
Affiliation(s)
- M Maurer
- Department of Dermatology and Allergy, Allergie-Centrum-Charité, Charité- Universitätsmedizin, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
|
25
|
Zhang X, Tan F, Brovkovych V, Zhang Y, Skidgel RA. Cross-talk between carboxypeptidase M and the kinin B1 receptor mediates a new mode of G protein-coupled receptor signaling. J Biol Chem 2011; 286:18547-61. [PMID: 21454694 DOI: 10.1074/jbc.m110.214940] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
G protein-coupled receptor (GPCR) signaling is affected by formation of GPCR homo- or heterodimers, but GPCR regulation by other cell surface proteins is not well understood. We reported that the kinin B1 receptor (B1R) heterodimerizes with membrane carboxypeptidase M (CPM), facilitating receptor signaling via CPM-mediated conversion of bradykinin or kallidin to des-Arg kinin B1R agonists. Here, we found that a catalytically inactive CPM mutant that still binds substrate (CPM-E264Q) also facilitates efficient B1R signaling by B2 receptor agonists bradykinin or kallidin. This response required co-expression of B1R and CPM-E264Q in the same cell, was disrupted by antibody that dissociates CPM from B1R, and was not found with a CPM-E264Q-B1R fusion protein. An additional mutation that reduced the affinity of CPM for C-terminal Arg and increased the affinity for C-terminal Lys inhibited the B1R response to bradykinin (with C-terminal Arg) but generated a response to Lys(9)-bradykinin. CPM-E264Q-mediated activation of B1Rs by bradykinin resulted in increased intramolecular fluorescence resonance energy transfer (FRET) in a B1R FRET construct, similar to that generated directly by a B1R agonist. In cytokine-treated human lung microvascular endothelial cells, disruption of B1R-CPM heterodimers inhibited B1R-dependent NO production stimulated by bradykinin and blocked the increased endothelial permeability caused by treatment with bradykinin and pyrogallol (a superoxide generator). Thus, CPM and B1Rs on cell membranes form a critical complex that potentiates B1R signaling. Kinin peptide binding to CPM causes a conformational change in the B1R leading to intracellular signaling and reveals a new mode of GPCR activation by a cell surface peptidase.
Collapse
Affiliation(s)
- Xianming Zhang
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois 60612, USA
| | | | | | | | | |
Collapse
|
26
|
Vasomotor and fibrinolytic responses to kinin receptor agonists in the atherosclerotic human lower limb. Heart Vessels 2011; 27:179-85. [DOI: 10.1007/s00380-011-0124-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 02/04/2011] [Indexed: 10/18/2022]
|
27
|
Spinetti G, Fortunato O, Cordella D, Portararo P, Kränkel N, Katare R, Sala-Newby GB, Richer C, Vincent MP, Alhenc-Gelas F, Tonolo G, Cherchi S, Emanueli C, Madeddu P. Tissue kallikrein is essential for invasive capacity of circulating proangiogenic cells. Circ Res 2010; 108:284-93. [PMID: 21164105 DOI: 10.1161/circresaha.110.236786] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
RATIONALE Homing of proangiogenic cells (PACs) is guided by chemoattractants and requires proteases to disrupt the extracellular matrix. The possibility that PAC recruitment involves an interaction between proteases and chemotactic factor receptors remains largely unexplored. OBJECTIVE To determine the role of human tissue kallikrein (hK1) in PAC invasion and its dependency on kinin receptor signaling. METHODS AND RESULTS Human mononuclear cells (MNCs) and culture-selected PACs express and release mature hK1 protein. HK1 gene (KLK1) silencing reduced PACs migratory, invasive, and proangiogenic activities. KLK1-knockout mouse bone marrow-derived MNCs showed similar impairments and were unable to support reparative angiogenesis in a mouse model of peripheral ischemia. Conversely, adenovirus-mediated KLK1 (Ad.KLK1) gene transfer enhanced PAC-associated functions, whereas the catalytically inactive variant R53H-KLK1 was ineffective. HK1-induced effects are mediated by a kinin B(2) receptor (B(2)R)-dependent mechanism involving inducible nitric oxide synthase and metalloproteinase-2 (MMP2). Lower hK1 protein levels were observed in PACs from type 2 diabetic (T2D) patients, whereas KLK1 mRNA levels were similar to those of healthy subjects, suggesting a post-transcriptional defect. Furthermore, B(2)R is normally expressed on T2D-PACs but remains uncoupled from downstream signaling. Importantly, whereas Ad.KLK1 alone could not restore T2D-PAC invasion capacity, combined KLK1 and B(2)R expression rescued the diabetic phenotype. CONCLUSIONS This study reveals new interactive components of the PACs invasive machinery, acting via protease- and kinin receptor-dependent mechanisms.
Collapse
Affiliation(s)
- Gaia Spinetti
- Chair of Experimental Cardiovascular Medicine, University of Bristol, Bristol BS28HW, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Bilgin M, Neuhof C, Doerr O, Benscheid U, Andrade SS, Most A, Abdallah Y, Parahuleva M, Guenduez D, Oliva ML, Erdogan A. Bauhinia bauhinioides cruzipain inhibitor reduces endothelial proliferation and induces an increase of the intracellular Ca2+ concentration. J Physiol Biochem 2010; 66:283-90. [DOI: 10.1007/s13105-010-0032-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Accepted: 06/30/2010] [Indexed: 10/19/2022]
|
29
|
Wende AR, Soto J, Olsen CD, Pires KMP, Schell JC, Larrieu-Lahargue F, Litwin SE, Kakoki M, Takahashi N, Smithies O, Abel ED. Loss of bradykinin signaling does not accelerate the development of cardiac dysfunction in type 1 diabetic akita mice. Endocrinology 2010; 151:3536-42. [PMID: 20501666 PMCID: PMC2940524 DOI: 10.1210/en.2010-0256] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Accepted: 04/27/2010] [Indexed: 12/11/2022]
Abstract
Bradykinin signaling has been proposed to play either protective or deleterious roles in the development of cardiac dysfunction in response to various pathological stimuli. To further define the role of bradykinin signaling in the diabetic heart, we examined cardiac function in mice with genetic ablation of both bradykinin B1 and B2 receptors (B1RB2R(-/-)) in the context of the Akita model of insulin-deficient type 1 diabetes (Ins2(Akita/+)). In 5-month-old diabetic and nondiabetic, wild-type and B1RB2R(-/-) mice, in vivo cardiac contractile function was determined by left-ventricular (LV) catheterization and echocardiography. Reactive oxygen species levels were measured by 2'-7'-dichlorofluorescein diacetate fluorescence. Mitochondrial function and ATP synthesis were determined in saponin-permeabilized cardiac fibers. LV systolic pressure and the peak rate of LV pressure rise and decline were decreased with diabetes but did not deteriorate further with loss of bradykinin signaling. Wall thinning and reduced ejection fractions in Akita mouse hearts were partially attenuated by B1RB2R deficiency, although other parameters of LV function were unaffected. Loss of bradykinin signaling did not increase fibrosis in Ins2(Akita/+) diabetic mouse hearts. Mitochondrial dysfunction was not exacerbated by B1RB2R deficiency, nor was there any additional increase in tissue levels of reactive oxygen species. Thus, loss of bradykinin B2 receptor signaling does not abrogate the previously reported beneficial effect of inhibition of B1 receptor signaling. In conclusion, complete loss of bradykinin expression does not worsen cardiac function or increase myocardial fibrosis in diabetes.
Collapse
MESH Headings
- Animals
- Bradykinin/metabolism
- Bradykinin/physiology
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/physiopathology
- Female
- Heart/physiopathology
- Heart Diseases/etiology
- Heart Diseases/genetics
- Heart Diseases/physiopathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Mitochondria, Heart/pathology
- Mitochondria, Heart/physiology
- Myocardium/pathology
- Oxidative Stress/genetics
- Receptor, Bradykinin B1/deficiency
- Receptor, Bradykinin B1/genetics
- Receptor, Bradykinin B2/deficiency
- Receptor, Bradykinin B2/genetics
- Signal Transduction/genetics
- Time Factors
Collapse
Affiliation(s)
- Adam R Wende
- Program in Molecular Medicine and Division of Endocrinology, Metabolism, and Diabetes, University of Utah, School of Medicine, Salt Lake City, Utah 84112, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Chao J, Shen B, Gao L, Xia CF, Bledsoe G, Chao L. Tissue kallikrein in cardiovascular, cerebrovascular and renal diseases and skin wound healing. Biol Chem 2010; 391:345-55. [PMID: 20180644 DOI: 10.1515/bc.2010.042] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Tissue kallikrein (KLK1) processes low-molecular weight kininogen to produce vasoactive kinins, which exert biological functions via kinin receptor signaling. Using various delivery approaches, we have demonstrated that tissue kallikrein through kinin B2 receptor signaling exhibits a wide spectrum of beneficial effects by reducing cardiac and renal injuries, restenosis and ischemic stroke, and by promoting angiogenesis and skin wound healing, independent of blood pressure reduction. Protection by tissue kallikrein in oxidative organ damage is attributed to the inhibition of apoptosis, inflammation, hypertrophy and fibrosis. Tissue kallikrein also enhances neovascularization in ischemic heart and limb. Moreover, tissue kallikrein/kinin infusion not only prevents but also reverses kidney injury, inflammation and fibrosis in salt-induced hypertensive rats. Furthermore, there is a wide time window for kallikrein administration in protection against ischemic brain infarction, as delayed kallikrein infusion for 24 h after cerebral ischemia in rats is effective in reducing neurological deficits, infarct size, apoptosis and inflammation. Importantly, in the clinical setting, human tissue kallikrein has been proven to be effective in the treatment of patients with acute brain infarction when injected within 48 h after stroke onset. Finally, kallikrein promotes skin wound healing and keratinocyte migration by direct activation of protease-activated receptor 1.
Collapse
Affiliation(s)
- Julie Chao
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, 29425, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Distinct kinin-induced functions are altered in circulating cells of young type 1 diabetic patients. PLoS One 2010; 5:e11146. [PMID: 20567501 PMCID: PMC2887352 DOI: 10.1371/journal.pone.0011146] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 05/19/2010] [Indexed: 01/06/2023] Open
Abstract
AIMS/HYPOTHESIS We aimed to understand early alterations in kinin-mediated migration of circulating angio-supportive cells and dysfunction of kinin-sensitive cells in type-1 diabetic (T1D) patients before the onset of cardiovascular disease. METHODS Total mononuclear cells (MNC) were isolated from peripheral blood of 28 T1D patients free from cardiovascular complications except mild background retinopathy (age: 34.8+/-1.6 years, HbA(1C): 7.9+/-0.2%) and 28 age- and sex-matched non-diabetic controls (H). We tested expression of kinin receptors by flow cytometry and migratory capacity of circulating monocytes and progenitor cells towards bradykinin (BK) in transwell migration assays. MNC migrating towards BK (BK(mig)) were assessed for capacity to support endothelial cell function in a matrigel assay, as well as generation of nitric oxide (NO) and superoxide (O(2) (-)*) by using the fluorescent probes diaminofluorescein and dihydroethidium. RESULTS CD14(hi)CD16(neg), CD14(hi)CD16(pos) and CD14(lo)CD16(pos) monocytes and circulating CD34(pos) progenitor cells did not differ between T1D and H subjects in their kinin receptor expression and migration towards BK. T1D BK(mig) failed to generate NO upon BK stimulation and supported endothelial cell network formation less efficiently than H BK(mig). In contrast, O(2) (-)* production was similar between groups. High glucose disturbed BK-induced NO generation by MNC-derived cultured angiogenic cells. CONCLUSIONS/INTERPRETATION Our data point out alterations in kinin-mediated functions of circulating MNC from T1D patients, occurring before manifest macrovascular damage or progressed microvascular disease. Functional defects of MNC recruited to the vessel wall might compromise endothelial maintenance, initially without actively promoting endothelial damage, but rather by lacking supportive contribution to endothelial regeneration and healing.
Collapse
|
32
|
Ban K, Kim KH, Cho CK, Sauvé M, Diamandis EP, Backx PH, Drucker DJ, Husain M. Glucagon-like peptide (GLP)-1(9-36)amide-mediated cytoprotection is blocked by exendin(9-39) yet does not require the known GLP-1 receptor. Endocrinology 2010; 151:1520-31. [PMID: 20172966 DOI: 10.1210/en.2009-1197] [Citation(s) in RCA: 182] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The widely expressed dipeptidyl peptidase-4 enzyme rapidly cleaves the gut hormone glucagon-like peptide-1 [GLP-1(7-36)amide] at the N terminus to generate GLP-1(9-36)amide. Both intact GLP-1(7-36)amide and GLP-1(9-36)amide exert cardioprotective actions in rodent hearts; however, the mechanisms underlying the actions of GLP-1(9-36)amide remain poorly understood. We used mass spectrometry of coronary effluents to demonstrate that isolated mouse hearts rapidly convert infused GLP-1(7-36)amide to GLP-1(9-36)amide. After ischemia-reperfusion (I/R) injury of isolated mouse hearts, administration of GLP-1(9-36)amide or exendin-4 improved functional recovery and reduced infarct size. The direct actions of these peptides were studied in cultured neonatal mouse cardiomyocytes. Both GLP-1(9-36)amide and exendin-4 increased levels of cAMP and phosphorylation of ERK1/2 and the phosphoinositide 3-kinase target protein kinase B/Akt. In I/R injury models in vitro, both peptides improved mouse cardiomyocyte viability and reduced lactate dehydrogenase release and caspase-3 activation. These effects were attenuated by inhibitors of ERK1/2 and phosphoinositide 3-kinase. Unexpectedly, the cardioprotective actions of GLP-1(9-36)amide were blocked by exendin(9-39) yet preserved in Glp1r(-/-) cardiomyocytes. Furthermore, GLP-1(9-36)amide, but not exendin-4, improved the survival of human aortic endothelial cells undergoing I/R injury, actions sensitive to the nitric oxide synthase inhibitor, N(G)-nitro-l-arginine methyl ester (L-NAME). In summary, our findings demonstrate separate actions for GLP-1(9-36)amide vs. the GLP-1R agonist exendin-4 and reveal the existence of a GLP-1(9-36)amide-responsive, exendin(9-39)-sensitive, cardioprotective signaling pathway distinct from that associated with the classical GLP-1 receptor.
Collapse
Affiliation(s)
- Kiwon Ban
- TMDT 3-904, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Liu X, Li Y, Liu Y, Luo Y, Wang D, Annex BH, Goldschmidt-Clermont PJ. Endothelial progenitor cells (EPCs) mobilized and activated by neurotrophic factors may contribute to pathologic neovascularization in diabetic retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 176:504-15. [PMID: 19948824 DOI: 10.2353/ajpath.2010.081152] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Diabetic retinopathy is characterized by pathological retinal neovascularization. Accumulating evidence has indicated that high levels of circulating endothelial progenitor cells (EPCs) are an important risk factor for neovascularization. Paradoxically, the reduction and dysfunction of circulating EPCs has been extensively reported in diabetic patients. We hypothesized that EPCs are differentially altered in the various vasculopathic complications of diabetes mellitus, exhibiting distinct behaviors in terms of angiogenic response to ischemia and growth factors and potentially playing a potent role in motivating vascular precursors to induce pathological neovascularization. Circulating levels of EPCs from diabetic retinopathy patients were analyzed by flow cytometry and by counting EPC colony-forming units, and serum levels of neurotrophic factors were measured by enzyme-linked immunosorbent assay. We found increased levels of nerve growth factor and brain-derived neurotrophic factor in the blood of diabetic retinopathy patients; this increase was correlated with the levels of circulating EPCs. In addition, we demonstrated that retinal cells released neurotrophic factors under hypoxic conditions to enhance EPC activity in vitro and to increase angiogenesis in a mouse ischemic hindlimb model. These results suggest that neurotrophic factors may induce neoangiogenesis through EPC activation, leading to the pathological retinal neovascularization. Thus, we propose that neovascularization in the ischemic retina might be regulated by overexpression of neurotrophic factors.
Collapse
Affiliation(s)
- Xialin Liu
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou 510060, China.
| | | | | | | | | | | | | |
Collapse
|
34
|
Majumder S, Gupta R, Reddy H, Sinha S, Muley A, Kolluru GK, Chatterjee S. Cadmium attenuates bradykinin-driven nitric oxide production by interplaying with the localization pattern of endothelial nitric oxide synthase. Biochem Cell Biol 2009; 87:605-20. [PMID: 19767824 DOI: 10.1139/o09-018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cadmium, a ubiquitous heavy metal, interferes with endothelial functions and angiogenesis. Bradykinin is a Ca-mobilizing soluble peptide that acts via nitric oxide to promote vasodilation and capillary permeability. The objective of the present study was to explore the Cd implications in bradykinin-dependent endothelial functions. An egg yolk angiogenesis model was employed to evaluate the effect of Cd on bradykinin-induced angiogenesis. The results demonstrate that 100 nmol/L Cd attenuated bradykinin-dependent angiogenesis. The results of the in vitro wound healing and tube formation assays by using EAhy 926, a transformed endothelial cell line, suggest that Cd blocked bradykinin-mediated endothelial migration and tube formation by 38% and 67%, respectively, while nitric oxide supplementation could reverse the effect of Cd on bradykinin-induced endothelial migration by 94%. The detection of nitric oxide by using a DAF-2DA fluorescent probe, Griess assay, and ultrasensitive electrode suggests that Cd blocked bradykinin-induced nitric oxide production. Fluorescence imaging of eNOS-GFP transfected endothelial cells, immunofluorescence, and Western blot studies of Cd and bradykinin-treated cells show that Cd interfered with the localization pattern of eNOS, which possibly attenuates nitric oxide production in part. Additionally, Ca imaging of Cd- and bradykinin-treated cells suggests that Cd blocked bradykinin-dependent Ca influx into the cells, thus partially blocking Ca-dependent nitric oxide production in endothelial cells. The results of this study conclude that Cd blunted the effect of bradykinin by interfering with the Ca-associated NOS activity specifically by impeding subcellular trafficking of eNOS.
Collapse
Affiliation(s)
- Syamantak Majumder
- Vascular Biology Lab, Life Sciences Division, AU-KBC Research Centre, MIT Campus, Anna University, Chennai 600044, Tamil Nadu, India
| | | | | | | | | | | | | |
Collapse
|
35
|
Westermann D, Walther T, Savvatis K, Escher F, Sobirey M, Riad A, Bader M, Schultheiss HP, Tschöpe C. Gene deletion of the kinin receptor B1 attenuates cardiac inflammation and fibrosis during the development of experimental diabetic cardiomyopathy. Diabetes 2009; 58:1373-81. [PMID: 19276445 PMCID: PMC2682670 DOI: 10.2337/db08-0329] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Diabetic cardiomyopathy is associated with increased mortality in patients with diabetes. The underlying pathology of this disease is still under discussion. We studied the role of the kinin B1 receptor on the development of experimental diabetic cardiomyopathy. RESEARCH DESIGN AND METHODS We utilized B1 receptor knockout mice and investigated cardiac inflammation, fibrosis, and oxidative stress after induction of streptozotocin (STZ)-induced diabetes. Furthermore, the left ventricular function was measured by pressure-volume loops after 8 weeks of diabetes. RESULTS B1 receptor knockout mice showed an attenuation of diabetic cardiomyopathy with improved systolic and diastolic function in comparison with diabetic control mice. This was associated with a decreased activation state of the mitogen-activated protein kinase p38, less oxidative stress, as well as normalized cardiac inflammation, shown by fewer invading cells and no increase in matrix metalloproteinase-9 as well as the chemokine CXCL-5. Furthermore, the profibrotic connective tissue growth factor was normalized, leading to a reduction in cardiac fibrosis despite severe hyperglycemia in mice lacking the B1 receptor. CONCLUSIONS These findings suggest that the B1 receptor is detrimental in diabetic cardiomyopathy in that it mediates inflammatory and fibrotic processes. These insights might have useful implications on future studies utilizing B1 receptor antagonists for treatment of human diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Dirk Westermann
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Thomas Walther
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Konstantinos Savvatis
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Felcicitas Escher
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Meike Sobirey
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Alexander Riad
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
| | - Heinz-Peter Schultheiss
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Carsten Tschöpe
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
- Corresponding author: Carsten Tschöpe,
| |
Collapse
|
36
|
Stone OA, Richer C, Emanueli C, van Weel V, Quax PHA, Katare R, Kraenkel N, Campagnolo P, Barcelos LS, Siragusa M, Sala-Newby GB, Baldessari D, Mione M, Vincent MP, Benest AV, Al Haj Zen A, Gonzalez J, Bates DO, Alhenc-Gelas F, Madeddu P. Critical role of tissue kallikrein in vessel formation and maturation: implications for therapeutic revascularization. Arterioscler Thromb Vasc Biol 2009; 29:657-64. [PMID: 19164804 PMCID: PMC2827862 DOI: 10.1161/atvbaha.108.182139] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Human Tissue Kallikrein (hKLK1) overexpression promotes an enduring neovascularization of ischemic tissue, yet the cellular mechanisms of hKLK1-induced arteriogenesis remain unknown. Furthermore, no previous study has compared the angiogenic potency of hKLK1, with its loss of function polymorphic variant, rs5515 (R53H), which possesses reduced kinin-forming activity. METHODS AND RESULTS Here, we demonstrate that tissue kallikrein knockout mice (KLK1-/-) show impaired muscle neovascularization in response to hindlimb ischemia. Gene-transfer of wild-type Ad.hKLK1 but not Ad.R53H-hKLK1 was able to rescue this defect. Similarly, in the rat mesenteric assay, Ad.hKLK1 induced a mature neovasculature with increased vessel diameter through kinin-B2 receptor-mediated recruitment of pericytes and vascular smooth muscle cells, whereas Ad.R53H-hKLK1 was ineffective. Moreover, hKLK1 but not R53H-hKLK1 overexpression in the zebrafish induced endothelial precursor cell migration and vascular remodeling. Furthermore, Ad.hKLK1 activates metalloproteinase (MMP) activity in normoperfused muscle and fails to promote reparative neovascularization in ischemic MMP9-/- mice, whereas its proarteriogenic action was preserved in ApoE-/- mice, an atherosclerotic model of impaired angiogenesis. CONCLUSIONS These results demonstrate the fundamental role of endogenous Tissue Kallikrein in vascular repair and provide novel information on the cellular and molecular mechanisms responsible for the robust arterialization induced by hKLK1 overexpression.
Collapse
Affiliation(s)
- Oliver A Stone
- Microvascular Research Laboratories, Bristol Heart Institute, Department of Physiology and Pharmacology, University of Bristol, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Bawolak MT, Gera L, Morissette G, Bouthillier J, Stewart JM, Gobeil LA, Lodge R, Adam A, Marceau F. Fluorescent ligands of the bradykinin B1 receptors: pharmacologic characterization and application to the study of agonist-induced receptor translocation and cell surface receptor expression. J Pharmacol Exp Ther 2009; 329:159-68. [PMID: 19136639 DOI: 10.1124/jpet.108.149724] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
Unlike the widely distributed and preformed B(2) receptors, the bradykinin B(1) receptors exhibit a highly regulated expression and minimal agonist-induced endocytosis. To evaluate the potential usefulness of fluorescent B(1) receptor probes applicable to live cell microscopy and cytofluorometry, combined chemical synthesis and pharmacologic evaluation have been conducted on novel 5(6)-carboxyfluorescein [5(6)CF]-containing peptides. Representative agents are the antagonist B-10376 [5(6)CF-epsilon-aminocaproyl-Lys-Lys-[Hyp(3), CpG(5), D-Tic(7), CpG(8)]des-Arg(9)-bradykinin] and the agonist B-10378 [5(6)CF-epsilon-aminocaproyl-Lys-des-Arg(9)-bradykinin]. B-10376 has a K(i) of 10 to 20 nM to displace [(3)H]Lys-des-Arg(9)-bradykinin from rabbit or human recombinant B(1) receptors expressed in human embryonic kidney (HEK) 293 cells and is a surmountable antagonist in the rabbit aorta contractility assay (pA(2), 7.49). B-10378 was a full agonist at the naturally expressed B(1) receptor (rabbit aorta contraction, calcium transients in human smooth muscle cells) and had a binding competition K(i) of 19 or 89 nM at the recombinant rabbit or human receptor, respectively. Both fluorescent probes can label with specificity human or rabbit B(1) receptors expressed in HEK 293 cells (epifluorescence or confocal microscopy), but the agonist was associated with discontinuous plasma membrane labeling, which coincided with that of a red-emitting caveolin-1 conjugate. Cytofluorometry with B-10376 was applied to recombinant and, in human vascular smooth muscle cells, to naturally expressed B(1) receptors. In all fluorescent applications, the specific labeling was reduced by an excess of a B(1) receptor nonpeptide antagonist. Despite the loss of affinity determined by the introduction of a fluorophore in B(1) receptor agonist or antagonist peptides, the resulting agents allow original applications (imaging in live cells, cytofluorometry).
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Binding, Competitive/drug effects
- Bradykinin/analogs & derivatives
- Bradykinin/pharmacology
- Calcium Signaling/drug effects
- Cell Line
- Cell Membrane/drug effects
- Cell Membrane/metabolism
- Fluorescent Dyes
- Humans
- Indicators and Reagents
- Ligands
- Microscopy, Fluorescence
- Muscle Contraction/drug effects
- Muscle Contraction/physiology
- Protein Transport/drug effects
- Rabbits
- Receptor, Bradykinin B1/agonists
- Receptor, Bradykinin B1/biosynthesis
- Receptor, Bradykinin B1/drug effects
- Receptors, Cell Surface/agonists
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/drug effects
Collapse
Affiliation(s)
- Marie-Thérèse Bawolak
- Centre de Recherche en Rhumatologie et Immunologie, Room T1-49, Centre Hospitalier Universitaire de Québec, Quebec, QC, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Côté J, Savard M, Bovenzi V, Bélanger S, Morin J, Neugebauer W, Larouche A, Dubuc C, Gobeil F. Novel kinin B1 receptor agonists with improved pharmacological profiles. Peptides 2009; 30:788-95. [PMID: 19150636 DOI: 10.1016/j.peptides.2008.12.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Revised: 12/17/2008] [Accepted: 12/18/2008] [Indexed: 10/21/2022]
Abstract
There is some evidence to suggest that inducible kinin B1 receptors (B1R) may play beneficial and protecting roles in cardiovascular-related pathologies such as hypertension, diabetes, and ischemic organ diseases. Peptide B1R agonists bearing optimized pharmacological features (high potency, selectivity and stability toward proteolysis) hold promise as valuable therapeutic agents in the treatment of these diseases. In the present study, we used solid-phase methodology to synthesize a series of novel peptide analogues based on the sequence of Sar[dPhe(8)]desArg(9)-bradykinin, a relatively stable peptide agonist with moderate affinity for the human B1R. We evaluated the pharmacological properties of these peptides using (1) in vitro competitive binding experiments on recombinant human B1R and B2R (for index of selectivity determination) in transiently transfected human embryonic kidney 293 cells (HEK-293T cells), (2) ex vivo vasomotor assays on isolated human umbilical veins expressing endogenous human B1R, and (3) in vivo blood pressure tests using anesthetized lipopolysaccharide-immunostimulated rabbits. Key chemical modifications at the N-terminus, the positions 3 and 5 on Sar[dPhe(8)]desArg(9)-bradykinin led to potent analogues. For example, peptides 18 (SarLys[Hyp(3),Cha(5), dPhe(8)]desArg(9)-bradykinin) and 20 (SarLys[Hyp(3),Igl(5), dPhe(8)]desArg(9)-bradykinin) outperformed the parental molecule in terms of affinity, functional potency and duration of action in vitro and in vivo. These selective agonists should be valuable in future animal and human studies to investigate the potential benefits of B1R activation.
Collapse
Affiliation(s)
- Jérôme Côté
- Department of Pharmacology, Université de Sherbrooke, Québec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Kränkel N, Katare RG, Siragusa M, Barcelos LS, Campagnolo P, Mangialardi G, Fortunato O, Spinetti G, Tran N, Zacharowski K, Wojakowski W, Mroz I, Herman A, Manning Fox JE, MacDonald PE, Schanstra JP, Bascands JL, Ascione R, Angelini G, Emanueli C, Madeddu P. Role of kinin B2 receptor signaling in the recruitment of circulating progenitor cells with neovascularization potential. Circ Res 2008; 103:1335-43. [PMID: 18927465 PMCID: PMC2821015 DOI: 10.1161/circresaha.108.179952] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Reduced migratory function of circulating angiogenic progenitor cells (CPCs) has been associated with impaired neovascularization in patients with cardiovascular disease (CVD). Previous findings underline the role of the kallikrein-kinin system in angiogenesis. We now demonstrate the involvement of the kinin B2 receptor (B(2)R) in the recruitment of CPCs to sites of ischemia and in their proangiogenic action. In healthy subjects, B(2)R was abundantly present on CD133(+) and CD34(+) CPCs as well as cultured endothelial progenitor cells (EPCs) derived from blood mononuclear cells (MNCs), whereas kinin B1 receptor expression was barely detectable. In transwell migration assays, bradykinin (BK) exerts a potent chemoattractant activity on CD133(+) and CD34(+) CPCs and EPCs via a B(2)R/phosphoinositide 3-kinase/eNOS-mediated mechanism. Migration toward BK was able to attract an MNC subpopulation enriched in CPCs with in vitro proangiogenic activity, as assessed by Matrigel assay. CPCs from cardiovascular disease patients showed low B(2)R levels and decreased migratory capacity toward BK. When injected systemically into wild-type mice with unilateral limb ischemia, bone marrow MNCs from syngenic B(2)R-deficient mice resulted in reduced homing of sca-1(+) and cKit(+)flk1(+) progenitors to ischemic muscles, impaired reparative neovascularization, and delayed perfusion recovery as compared with wild-type MNCs. Similarly, blockade of the B(2)R by systemic administration of icatibant prevented the beneficial effect of bone marrow MNC transplantation. BK-induced migration represents a novel mechanism mediating homing of circulating angiogenic progenitors. Reduction of BK sensitivity in progenitor cells from cardiovascular disease patients might contribute to impaired neovascularization after ischemic complications.
Collapse
Affiliation(s)
- Nicolle Kränkel
- Experimental Cardiovascular Medicine, Bristol Heart Institute, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wang PHM, Cenedeze MA, Campanholle G, Malheiros DMAC, Torres HADM, Pesquero JB, Pacheco-Silva A, Câmara NOS. Deletion of bradykinin B1 receptor reduces renal fibrosis. Int Immunopharmacol 2008; 9:653-7. [PMID: 19010452 DOI: 10.1016/j.intimp.2008.10.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Accepted: 10/27/2008] [Indexed: 02/06/2023]
Abstract
The Kallikrein-kinin system works through activation of two receptors. One constitutive, named B2 receptor (B2R) and another inducible, denominated B1 receptor (B1R). In renal fibrosis, B2R receptor activation appears to be protective, however B1R participation is unveiled. The aim of this study was to analyze how the deletion of the B1R would modify tissue responses after unilateral ureteral obstruction (UUO). For that, B1R knockout (B1KO) and wild-type mice (B1B2WT) were subjected to UUO and sacrificed at days 1, 5 and 14. Renal dysfunction was assayed by urine proteinuria/creatinine ratio and percentage of tubulointerstitial fibrosis. Kidneys were harvested at day 5 to analyze anti and pro-inflammatory molecules expression by real-time PCR. We demonstrated that at all time points, B1KO mice presented lower proteinuria/creatinine ratio from bladder urine. B1KO protection was reinforced by its lower tubular interstitial fibrosis percentage at day 14 (B1B2WT: 12.16+/-1.53% vs. B1KO: 6.73+/-1.07%, p<0.02). UUO was able to induce B1R expression and its highest transcription was achieved at day 5. At this day, B1KO had significant lower expression of pro-inflammatory molecules such as TGF-beta, MCP-1, OPN and IL-6 and higher anti-inflammatory components, as IL-10 and HO-1. Herein, we observed that B1R deletion may be an important component in renal fibrosis prevention.
Collapse
Affiliation(s)
- Pamella Huey Mei Wang
- Laboratório de Imunologia Clínica e Experimental, Division of Nephrology, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Molecular mechanism of limbsʼ postischemic revascularization improved by perindopril in diabetic rats. Chin Med J (Engl) 2008. [DOI: 10.1097/00029330-200811010-00004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
42
|
Hillmeister P, Lehmann KE, Bondke A, Witt H, Duelsner A, Gruber C, Busch HJ, Jankowski J, Ruiz-Noppinger P, Hossmann KA, Buschmann IR. Induction of cerebral arteriogenesis leads to early-phase expression of protease inhibitors in growing collaterals of the brain. J Cereb Blood Flow Metab 2008; 28:1811-23. [PMID: 18594555 DOI: 10.1038/jcbfm.2008.69] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cerebral arteriogenesis constitutes a promising therapeutic concept for cerebrovascular ischaemia; however, transcriptional profiles important for therapeutic target identification have not yet been investigated. This study aims at a comprehensive characterization of transcriptional and morphologic activation during early-phase collateral vessel growth in a rat model of adaptive cerebral arteriogenesis. Arteriogenesis was induced using a three-vessel occlusion (3-VO) rat model of nonischaemic cerebral hypoperfusion. Collateral tissue from growing posterior cerebral artery (PCA) and posterior communicating artery (Pcom) was selectively isolated avoiding contamination with adjacent tissue. We detected differential gene expression 24 h after 3-VO with 164 genes significantly deregulated. Expression patterns contained gene transcripts predominantly involved in proliferation, inflammation, and migration. By using scanning electron microscopy, morphologic activation of the PCA endothelium was detected. Furthermore, the PCA showed induced proliferation (PCNA staining) and CD68+ macrophage staining 24 h after 3-VO, resulting in a significant increase in diameter within 7 days after 3-VO, confirming the arteriogenic phenotype. Analysis of molecular annotations and networks associated with differentially expressed genes revealed that early-phase cerebral arteriogenesis is characterised by the expression of protease inhibitors. These results were confirmed by quantitative real-time reverse transcription-PCR, and in situ hybridisation localised the expression of tissue inhibitor of metalloproteinase-1 (TIMP-1) and kininogen to collateral arteries, showing that TIMP-1 and kininogen might be molecular markers for early-phase cerebral arteriogenesis.
Collapse
Affiliation(s)
- Philipp Hillmeister
- Research Group for Experimental and Clinical Arteriogenesis, Department of Internal Medicine/Cardiology, (CC13), CCR-Center for Cardiovascular Research, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Westermann D, Lettau O, Sobirey M, Riad A, Bader M, Schultheiss HP, Tschöpe C. Doxorubicin cardiomyopathy-induced inflammation and apoptosis are attenuated by gene deletion of the kinin B1 receptor. Biol Chem 2008; 389:713-8. [PMID: 18627295 DOI: 10.1515/bc.2008.070] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Clinical use of the anthracycline doxorubicin (DOX) is limited by its cardiotoxic effects, which are attributed to the induction of apoptosis. To elucidate the possible role of the kinin B1 receptor (B1R) during the development of DOX cardiomyopathy, we studied B1R knockout mice (B1R(-/-)) by investigating cardiac inflammation and apoptosis after induction of DOX-induced cardiomyopathy. DOX control mice showed cardiac dysfunction measured by pressure-volume loops in vivo. This was associated with a reduced activation state of AKT, as well as an increased bax/bcl2 ratio in Western blots, indicating cardiac apoptosis. Furthermore, mRNA levels of the proinflammatory cytokine interleukin 6 were increased in the cardiac tissue. In DOX B1R(-/-) mice, cardiac dysfunction was improved compared to DOX control mice, which was associated with normalization of the bax/bcl-2 ratio and interleukin 6, as well as AKT activation state. These findings suggest that B1R is detrimental in DOX cardiomyopathy in that it mediates the inflammatory response and apoptosis. These insights might have useful implications for future studies utilizing B1R antagonists for treatment of human DOX cardiomyopathy.
Collapse
Affiliation(s)
- Dirk Westermann
- Charité-Universitätsmedizin Berlin, Department of Cardiology and Pneumology, Campus Benjamin Franklin, D-12200 Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
44
|
Wang PHM, Campanholle G, Cenedeze MA, Feitoza CQ, Gonçalves GM, Landgraf RG, Jancar S, Pesquero JB, Pacheco-Silva A, Câmara NOS. Bradykinin [corrected] B1 receptor antagonism is beneficial in renal ischemia-reperfusion injury. PLoS One 2008; 3:e3050. [PMID: 18725957 PMCID: PMC2516176 DOI: 10.1371/journal.pone.0003050] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Accepted: 07/31/2008] [Indexed: 02/07/2023] Open
Abstract
Previously we have demonstrated that bradykinin B1 receptor deficient mice (B1KO) were protected against renal ischemia and reperfusion injury (IRI). Here, we aimed to analyze the effect of B1 antagonism on renal IRI and to study whether B1R knockout or antagonism could modulate the renal expression of pro and anti-inflammatory molecules. To this end, mice were subjected to 45 minutes ischemia and reperfused at 4, 24, 48 and 120 hours. Wild-type mice were treated intra-peritoneally with antagonists of either B1 (R-954, 200 µg/kg) or B2 receptor (HOE140, 200 µg/kg) 30 minutes prior to ischemia. Blood samples were collected to ascertain serum creatinine level, and kidneys were harvested for gene transcript analyses by real-time PCR. Herein, B1R antagonism (R-954) was able to decrease serum creatinine levels, whereas B2R antagonism had no effect. The protection seen under B1R deletion or antagonism was associated with an increased expression of GATA-3, IL-4 and IL-10 and a decreased T-bet and IL-1β transcription. Moreover, treatment with R-954 resulted in lower MCP-1, and higher HO-1 expression. Our results demonstrated that bradykinin B1R antagonism is beneficial in renal IRI.
Collapse
Affiliation(s)
- Pamella H. M. Wang
- Laboratório de Imunologia Clínica e Experimental, Division of Nephrology, Universidade Federal de São Paulo, São Paulo, Brazil
- * E-mail:
| | - Gabriela Campanholle
- Laboratório de Imunobiologia de Transplantes, Department of Immunology, Universidade de São Paulo, São Paulo, Brazil
| | - Marcos A. Cenedeze
- Laboratório de Imunologia Clínica e Experimental, Division of Nephrology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Carla Q. Feitoza
- Laboratório de Imunologia Clínica e Experimental, Division of Nephrology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Giselle M. Gonçalves
- Laboratório de Imunologia Clínica e Experimental, Division of Nephrology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Richardt G. Landgraf
- Laboratório de Imunofarmacologia, Department of Immunology, Universidade de São Paulo, São Paulo, Brazil
| | - Sonia Jancar
- Laboratório de Imunofarmacologia, Department of Immunology, Universidade de São Paulo, São Paulo, Brazil
| | - João B. Pesquero
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Alvaro Pacheco-Silva
- Laboratório de Imunologia Clínica e Experimental, Division of Nephrology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Niels O. S. Câmara
- Laboratório de Imunologia Clínica e Experimental, Division of Nephrology, Universidade Federal de São Paulo, São Paulo, Brazil
- Laboratório de Imunobiologia de Transplantes, Department of Immunology, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
45
|
Wright JK, Botha JH, Naidoo S. Influence of the kallikrein-kinin system on prostate and breast tumour angiogenesis. Tumour Biol 2008; 29:130-6. [PMID: 18577888 DOI: 10.1159/000141918] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Accepted: 04/18/2008] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Angiogenesis is important for the growth and progression of cancer cells. There is some evidence that the kallikrein-kinin system (KKS) is involved in cancer and angiogenesis. The present study investigated the effect of increasing concentrations of prostate and breast tumour cell metabolites on the proliferation of cultured endothelial cells, their tissue kallikrein (TK) secretion and KKS expression. METHODS Expression of TK and kinin receptors was investigated by immunochemistry, and secretion of TK by ELISA. Cell proliferation was measured by a chromogenic assay. KKS proteins were also immunolocalised in an endothelial tumour co-culture model. RESULTS KKS proteins were found in projections of all cell types as well as at points of heterogeneous contact. Tumour metabolites increased the secretion of TK from endothelial cells, with corresponding decreases in intracellular amounts, while also increasing proliferation of the endothelial cells. CONCLUSIONS These findings indicate that the KKS may be one of the more important players in angiogenesis associated with prostate and breast tumours.
Collapse
Affiliation(s)
- Jaclyn K Wright
- Department of Therapeutics and Medicines Management, Nelson R Mandela School of Medicine Durban, South Africa
| | | | | |
Collapse
|
46
|
Smith RS, Gao L, Chao L, Chao J. Tissue kallikrein and kinin infusion promotes neovascularization in limb ischemia. Biol Chem 2008; 389:725-30. [DOI: 10.1515/bc.2008.084] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Adenovirus-mediated kallikrein delivery has been shown to promote blood vessel growth in the limb under both ischemic and normoperfused conditions. Here we investigated whether a continuous supply of kallikrein and kinin peptide can induce neovascularization in a rat model of hindlimb ischemia. Rats underwent femoral artery ligation and localized injection of tissue kallikrein, bradykinin or B1 receptor agonist, followed by infusion of proteins by osmotic minipump. Regional blood flow was monitored weekly by laser Doppler perfusion imaging. Three weeks after surgery, rats receiving kallikrein and kinins showed a significant increase in the perfusion ratio of ischemic vs. normoperfused limb compared to control rats. Similarly, a microsphere assay showed that kallikrein and kinins significantly increased regional blood flow without altering blood pressure. Moreover, kallikrein and kinins significantly augmented capillary and arteriole densities, as quantified by immunostaining with CD-31 and smooth muscle α-actin. Both tissue kallikrein and bradykinin increased hemoglobin content in Matrigel implants in mice, providing further evidence of the angiogenic properties. Kinins, when delivered subcutaneously via Matrigel in rats, also increased regional perfusion. This is the first demonstration that local application of tissue kallikrein protein or kinin peptide has therapeutic value in the treatment of ischemic disease by promoting neovascularization.
Collapse
|
47
|
Merino VF, Todiras M, Campos LA, Saul V, Popova E, Baltatu OC, Pesquero JB, Bader M. Increased susceptibility to endotoxic shock in transgenic rats with endothelial overexpression of kinin B(1) receptors. J Mol Med (Berl) 2008; 86:791-8. [PMID: 18425495 DOI: 10.1007/s00109-008-0345-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2007] [Revised: 01/17/2008] [Accepted: 01/30/2008] [Indexed: 02/06/2023]
Abstract
Two kinin receptors have been described, the inducible B(1) and the constitutive B(2). B(1) receptors are important in cardiovascular homeostasis and inflammation. To further clarify their vascular function, we have generated transgenic rats (TGR(Tie2B(1))) overexpressing the B(1) receptor exclusively in the endothelium. Endothelial cell-specific expression was confirmed by B(1)-agonist-induced relaxation of isolated aorta, which was abolished by endothelial denudation of the vessel. This vasodilatation was mediated by nitric oxide (NO) and K(+) channels. TGR(Tie2B(1)) rats were normotensive but, in contrast to controls, reacted with a marked fall in blood pressure and increased vascular permeability after intravenous injection of a B(1) agonist. After lipopolysaccharide treatment, they present a more pronounced hypotensive response and marked bradycardia associated with increased mortality when compared to non-transgenic control animals. Thus, the transgenic rats overexpressing kinin B(1) receptors exclusively in the endothelium generated in this study support an important role of this receptor in the vasculature during the pathogenesis of endotoxic shock.
Collapse
Affiliation(s)
- Vanessa F Merino
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, Berlin-Buch, Berlin, 13125, Germany
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Diabetic retinopathy and diabetic nephropathy are common microvascular complications of diabetes. The kallikrein-kinin system (KKS) has been implicated in the development of both conditions, and, in particular, bradykinin and its receptors have been shown to exert angiogenic and proinflammatory actions. Several of the key processes that underlie the development of diabetic retinopathy, such as increased vascular permeability, edema, neovascularization, and inflammatory changes, have been associated with the KKS, and recent work has shown that components of the KKS, including plasma kallikrein, factor XIIa, and high-molecular-weight kininogen, are present in the vitreous of people with diabetic retinopathy. The role of the KKS in the development of diabetic nephropathy is controversial, with both adverse and protective effects of bradykinin and its receptors reported. The review examines the role of the KKS in pathways central to the development of diabetic retinopathy and compares this with reported actions of this system in diabetic nephropathy. The possibility of therapeutic intervention targeting bradykinin and its receptors as treatment for diabetic microvascular conditions is considered.
Collapse
|
49
|
Essential role of TM V and VI for binding the C-terminal sequences of Des-Arg-kinins. Int Immunopharmacol 2008; 8:282-8. [DOI: 10.1016/j.intimp.2007.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2007] [Revised: 08/31/2007] [Accepted: 09/02/2007] [Indexed: 11/20/2022]
|
50
|
Westermann D, Schultheiss HP, Tschöpe C. New perspective on the tissue kallikrein–kinin system in myocardial infarction: Role of angiogenesis and cardiac regeneration. Int Immunopharmacol 2008; 8:148-54. [DOI: 10.1016/j.intimp.2007.07.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2007] [Revised: 07/19/2007] [Accepted: 07/23/2007] [Indexed: 11/17/2022]
|