1
|
Lu L, Li DX, Chen W, Li GS, Hao P. Bradykinin-(1-9) mitigates autophagy through upregulating PI3K/Akt in rats with myocardial infarction. Biochem Biophys Res Commun 2023; 660:35-42. [PMID: 37060829 DOI: 10.1016/j.bbrc.2023.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/26/2023] [Accepted: 04/04/2023] [Indexed: 04/17/2023]
Abstract
The cardioprotective mechanisms of bradykinin-(1-9) in myocardial infarction were unclear. We investigated the effect of bradykinin-(1-9) on cardiac function, fibrosis, and autophagy induced by myocardial infarction and identified the mechanisms involved. To investigate the cardioprotective effect of bradykinin-(1-9), various doses of bradykinin-(1-9), its B2 receptor blocker HOE140, or their combination were administered to rats via subcutaneous osmotic minipump implantation before myocardial infarction. After 2 days, myocardial infarction was induced by ligation of the left anterior descending coronary artery. After 2 weeks, echocardiographic measurements and euthanasia were performed. Bradykinin-(1-9) treatment attenuated left ventricular dysfunction, fibrosis, and autophagy in rats with myocardial infarction, which was partially reversed by HOE140 administration. Moreover, the downregulatory effect of bradykinin-(1-9) on autophagy was partially reversed by combination with the PI3K inhibitor LY294002. Thus, bradykinin-(1-9) inhibits myocardial infarction-induced cardiomyocyte autophagy by upregulating the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Lin Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China; Department of Cardiovascular Medicine, the Third Affiliated Hospital of Shandong First Medical University, Jinan, 250031, Shandong Province, China
| | - Dai-Xu Li
- Department of Cardiovascular Medicine, The Fourth People's Hospital of Jinan, Jinan, 250031, Shandong Province, China
| | - Wei Chen
- Department of Cardiovascular Medicine, the Third Affiliated Hospital of Shandong First Medical University, Jinan, 250031, Shandong Province, China
| | - Gui-Shuang Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China.
| | - Panpan Hao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China.
| |
Collapse
|
2
|
Song J, Du J, Tan X, Wu Z, Yuan J, Cong B. Dexmedetomidine protects the heart against ischemia reperfusion injury via regulation of the bradykinin receptors. Eur J Pharmacol 2021; 911:174493. [PMID: 34506777 DOI: 10.1016/j.ejphar.2021.174493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/26/2021] [Accepted: 09/06/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Dexmedetomidine (DEX) has been reported to protect the heart against ischemia reperfusion (I/R) injury. However, the exact mechanisms are still not fully understood. METHODS AND RESULTS A rat cardiac I/R injury model was induced by ligation of the left anterior descending coronary artery for 1 h and subsequent reperfusion for 2 h, and DEX was administered intravenously 30 min before ischemia. We confirmed that DEX treatment mitigated cardiac I/R injury. Interestingly, we found that DEX regulated the expression of bradykinin (BK) receptors (B1R and B2R) in rat hearts during I/R injury and enhanced the protective action of BK administered during reperfusion. Moreover, in vitro hypoxia reoxygenation (H/R) injury was induced in neonatal rat cardiomyocytes (CMs), and DEX was administered 1 h before hypoxia. The in vitro findings were consistent with the in vivo experiments. We found that an α2-adrenoceptor (α2-AR) antagonist (yohimbine) completely aborted DEX-induced B1R and B2R regulation; an adenylyl cyclase (AC) agonist (forskolin) blocked B1R downregulation, while a phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002) blocked B2R upregulation. The above findings indicated that DEX interacted with α2-AR in cardiomyocytes, inhibited B1R expression via suppression of AC, and stimulated B2R expression via activation of PI3K. CONCLUSIONS DEX regulates BK receptor expression and potentiates the protection of BK in cardiac I/R injury, which suggests that modulating endogenous cardioprotective factors may play an important role in DEX-induced cardioprotection.
Collapse
Affiliation(s)
- Jinchao Song
- Department of Anesthesiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, China.
| | - Jiankui Du
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Xing Tan
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Zhaotang Wu
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Jihong Yuan
- Department of Nephropathy, Shanghai Seventh People's Hospital, Shanghai, 200137, China
| | - Binhai Cong
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China.
| |
Collapse
|
3
|
Wu C, Zhou XX, Li JZ, Qiang HF, Wang Y, Li G. Pretreatment of cardiac progenitor cells with bradykinin attenuates H 2O 2-induced cell apoptosis and improves cardiac function in rats by regulating autophagy. Stem Cell Res Ther 2021; 12:437. [PMID: 34353364 PMCID: PMC8340370 DOI: 10.1186/s13287-021-02503-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 07/01/2021] [Indexed: 02/07/2023] Open
Abstract
Background Previous studies have demonstrated that human cardiac c-Kit+ progenitor cells (hCPCs) can effectively improve ischemic heart disease. However, the major challenge in applying hCPCs to clinical therapy is the low survival rate of graft hCPCs in the host heart, which limited the benefit of transplanted hCPCs. Bradykinin (BK) is a principal active agent of the tissue kinin-kallikrein system. Our previous studies have highlighted that BK mediated the growth and migration of CPCs by regulating Ca2+ influx. However, the protective effect of BK on CPCs, improvement in the survival rate of BK-pretreated hCPCs in the infarcted heart, and the related mechanism remain elusive. Methods HCPCs were treated with H2O2 to induce cell apoptosis and autophagy, and different concentration of BK was applied to rescue the H2O2-induced injury detected by MTT assay, TUNEL staining, flow cytometry, western blotting, and mitoSOX assays. The role of autophagy in the anti-apoptotic effect of BK was chemically activated or inhibited using the autophagy inducer, rapamycin, or the inhibitor, 3-methyladenine (3-MA). To explore the protective effect of BK on hCPCs, 3-MA or BK-pretreated hCPCs were transplanted into the myocardial infarcted rats. An echocardiogram was used to determine cardiac function, H&E and Masson staining were employed to assess pathological characteristics, HLA gene expression was quantified by qRT-PCR, and immunostaining was applied to examine neovascularization using confocal microscopy. Results The in vitro results showed that BK suppressed H2O2-induced hCPCs apoptosis and ROS production in a concentration-dependent manner by promoting pAkt and Bcl-2 expression and reducing cleaved caspase 3 and Bax expression. Moreover, BK restrained the H2O2-induced cell autophagy by decreasing LC3II/I, Beclin1, and ATG5 expression and increasing P62 expression. In the in vivo experiment, the transplanted BK- or 3-MA-treated hCPCs were found to be more effectively improved cardiac function by decreasing cardiomyocyte apoptosis, inflammatory infiltration, and myocardial fibrosis, and promoting neovascularization in the infarcted heart, compared to untreated-hCPCs or c-kit- cardiomyocytes (CPC- cells). Conclusions Our present study established a new method to rescue transplanted hCPCs in the infarcted cardiac area via regulating cell apoptosis and autophagy of hCPCs by pretreatment with BK, providing a new therapeutic option for heart failure. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02503-6.
Collapse
Affiliation(s)
- Chan Wu
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, 361015, Fujian, China
| | - Xiao-Xia Zhou
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, 361015, Fujian, China
| | - Jing-Zhou Li
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, 361015, Fujian, China
| | - Hai-Feng Qiang
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, 361015, Fujian, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, 361015, Fujian, China.
| | - Gang Li
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, 361015, Fujian, China.
| |
Collapse
|
4
|
Hamid S, Rhaleb IA, Kassem KM, Rhaleb NE. Role of Kinins in Hypertension and Heart Failure. Pharmaceuticals (Basel) 2020; 13:E347. [PMID: 33126450 PMCID: PMC7692223 DOI: 10.3390/ph13110347] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
The kallikrein-kinin system (KKS) is proposed to act as a counter regulatory system against the vasopressor hormonal systems such as the renin-angiotensin system (RAS), aldosterone, and catecholamines. Evidence exists that supports the idea that the KKS is not only critical to blood pressure but may also oppose target organ damage. Kinins are generated from kininogens by tissue and plasma kallikreins. The putative role of kinins in the pathogenesis of hypertension is discussed based on human mutation cases on the KKS or rats with spontaneous mutation in the kininogen gene sequence and mouse models in which the gene expressing only one of the components of the KKS has been deleted or over-expressed. Some of the effects of kinins are mediated via activation of the B2 and/or B1 receptor and downstream signaling such as eicosanoids, nitric oxide (NO), endothelium-derived hyperpolarizing factor (EDHF) and/or tissue plasminogen activator (T-PA). The role of kinins in blood pressure regulation at normal or under hypertension conditions remains debatable due to contradictory reports from various laboratories. Nevertheless, published reports are consistent on the protective and mediating roles of kinins against ischemia and cardiac preconditioning; reports also demonstrate the roles of kinins in the cardiovascular protective effects of the angiotensin-converting enzyme (ACE) and angiotensin type 1 receptor blockers (ARBs).
Collapse
Affiliation(s)
- Suhail Hamid
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA; (S.H.); (I.A.R.)
| | - Imane A. Rhaleb
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA; (S.H.); (I.A.R.)
| | - Kamal M. Kassem
- Division of Cardiology, Department of Internal Medicine, University of Louisville Medical Center, Louisville, KY 40202, USA;
| | - Nour-Eddine Rhaleb
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA; (S.H.); (I.A.R.)
- Department of Physiology, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
5
|
Expression of B2 Receptor on Circulating CD34-Positive Cells and Outcomes of Myocardial Infarction. DISEASE MARKERS 2019; 2019:7816438. [PMID: 31360266 PMCID: PMC6644252 DOI: 10.1155/2019/7816438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/12/2019] [Accepted: 04/16/2019] [Indexed: 11/17/2022]
Abstract
Background Bradykinin B2 receptor (B2R) is a widely expressed cell surface receptor. The relationship between B2R expression on circulating CD34+ cells and prognosis of myocardial infarction remains unknown. Methods We analyzed the expression of B2R on circulating CD34-positive cells and plasma VEGF concentration in 174 myocardial infarction patients. All involved patients were divided into two groups: high B2R group and low B2R group according to the median B2R expression percentage. 48 months of follow-up was performed. The endpoints were heart failure and revascularization. Results The plasma level of VEGF in the low B2R group is 67 ± 12 pg/mL, whereas the high B2R group has significantly elevated VEGF levels of 145 ± 27 pg/mL (P < 0.001). The concentration of VEGF has correlated with expression of B2R (r = 0.574, P < 0.001). During the 48 months of follow-up, low expression of B2 receptor on circulating CD34-positive cells indicates the high incidence of heart failure (hazard ratio: 2.247; 95% confidence interval: 1.110-4.547; P = 0.024) and revascularization (hazard ratio: 2.335; 95% confidence interval: 1.075-5.074; P = 0.032). Kaplan-Meier survival analysis showed that the cumulative hazard of heart failure (P = 0.014) and revascularization (P = 0.032) has significant differences between low B2R and high B2R. Conclusion Low expression of B2R on circulating progenitor cells indicated the poor outcomes of myocardial infarction.
Collapse
|
6
|
Arichi S, Sasaki-Hamada S, Kadoya Y, Ogata M, Ishibashi H. Excitatory effect of bradykinin on intrinsic neurons of the rat heart. Neuropeptides 2019; 75:65-74. [PMID: 31047706 DOI: 10.1016/j.npep.2019.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/07/2019] [Accepted: 04/23/2019] [Indexed: 01/16/2023]
Abstract
The heart receives sympathetic and parasympathetic innervation through the intrinsic cardiac nervous system. Although bradykinin (BK) has negative inotropic and chronotropic properties of cardiac contraction, the direct effect of BK on the intrinsic neural network of the heart is still unclear. In the present study, the effect of BK on the intracardiac ganglion neurons isolated from rats was investigated using the perforated patch-clamp technique. Under current-clamp conditions, application of 0.1 μM BK depolarized the membrane, accompanied by repetitive firing of action potentials. When BK was applied repeatedly, the second responses were considerably less intense than the first application. The BK action was fully inhibited by the B2 receptor antagonist Hoe-140, but not by the B1 receptor antagonist des-Arg9-[Leu8]-BK. The BK response was mimicked by the B2 agonist [Hyp3]-BK. The BK-induced depolarization was inhibited by the phospholipase C inhibitor U-73122. BK evoked inward currents under voltage-clamp conditions at a holding potential of -60 mV. Removal of extracellular Ca2+ markedly increased the BK-induced currents, suggesting an involvement of Ca2+-permeable non-selective cation channels. The muscarinic agonist oxotremorine-M (OxoM) also elicited the extracellular Ca2+-sensitive cationic currents. The OxoM response did not exhibit rundown with repeated agonist application. The amplitude of current evoked by 1 μM OxoM was comparable to that induced by 0.1 μM BK. Co-application of 0.1 μM BK and 1 μM OxoM elicited the current whose peak amplitude was almost the same as that elicited by OxoM alone, suggesting that BK and OxoM activate same cation channels. BK also reduced the amplitude of M-current, while the M-current inhibitor XE-991 affected neither resting membrane potential nor the BK-induced depolarization. From these results, we suggest that BK regulates excitability of intrinsic cardiac neurons by both an activation of non-selective cation channels and an inhibition of M-type K+ channels through B2 receptors.
Collapse
Affiliation(s)
- Shiho Arichi
- Department of Physiology, School of Allied Health Sciences, Kitasato University, Sagamihara 252-0373, Japan; Department of Brain Science, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan
| | - Sachie Sasaki-Hamada
- Department of Physiology, School of Allied Health Sciences, Kitasato University, Sagamihara 252-0373, Japan
| | - Yuichi Kadoya
- Department of Anatomical Science, School of Allied Health Sciences, Kitasato University, Sagamihara 252-0373, Japan
| | - Masanori Ogata
- Department of Physiology, School of Allied Health Sciences, Kitasato University, Sagamihara 252-0373, Japan; Department of Brain Science, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan
| | - Hitoshi Ishibashi
- Department of Physiology, School of Allied Health Sciences, Kitasato University, Sagamihara 252-0373, Japan; Department of Brain Science, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan.
| |
Collapse
|
7
|
Brailoiu E, McGuire M, Shuler SA, Deliu E, Barr JL, Abood ME, Brailoiu GC. Modulation of cardiac vagal tone by bradykinin acting on nucleus ambiguus. Neuroscience 2017; 365:23-32. [PMID: 28951324 DOI: 10.1016/j.neuroscience.2017.09.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 09/11/2017] [Accepted: 09/18/2017] [Indexed: 12/21/2022]
Abstract
Bradykinin (BK), a component of the kallikrein-kininogen-kinin system exerts multiple effects via B1 and B2 receptor activation. In the cardiovascular system, bradykinin has cardioprotective and vasodilator properties. We investigated the effect of BK on cardiac-projecting neurons of nucleus ambiguus, a key site for the parasympathetic cardiac regulation. BK produced a dose-dependent increase in cytosolic Ca2+ concentration. Pretreatment with HOE140, a B2 receptor antagonist, but not with R715, a B1 receptor antagonist, abolished the response to BK. A selective B2 receptor agonist, but not a B1 receptor agonist, elicited an increase in cytosolic Ca2+ similarly to BK. Inhibition of N-type voltage-gated Ca2+ channels with ω-conotoxin GVIA had no effect on the Ca2+ signal produced by BK, while pretreatment with ω-conotoxin MVIIC, a blocker of P/Q-type of Ca2+ channels, significantly diminished the effect of BK. Pretreatment with xestospongin C and 2-aminoethoxydiphenyl borate, antagonists of inositol 1,4,5-trisphosphate receptors, abolished the response to BK. Inhibition of ryanodine receptors reduced the BK-induced Ca2+ increase, while disruption of lysosomal Ca2+ stores with bafilomycin A1 did not affect the response. BK produced a dose-dependent depolarization of nucleus ambiguus neurons, which was prevented by the B2 receptor antagonist. In vivo studies indicate that microinjection of BK into nucleus ambiguus elicited bradycardia in conscious rats via B2 receptors. In summary, in cardiac vagal neurons of nucleus ambiguus, BK activates B2 receptors promoting Ca2+ influx and Ca2+ release from endoplasmic reticulum, and membrane depolarization; these effects are translated in vivo by bradycardia.
Collapse
Affiliation(s)
- Eugen Brailoiu
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Philadelphia, PA 19140, United States
| | - Matthew McGuire
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Philadelphia, PA 19107, United States
| | - Shadaria A Shuler
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Philadelphia, PA 19107, United States
| | - Elena Deliu
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Philadelphia, PA 19140, United States
| | - Jeffrey L Barr
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Philadelphia, PA 19140, United States
| | - Mary E Abood
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Philadelphia, PA 19140, United States
| | - G Cristina Brailoiu
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Philadelphia, PA 19107, United States.
| |
Collapse
|
8
|
Nguyen NT, Lindsey ML, Jin YF. Systems analysis of gene ontology and biological pathways involved in post-myocardial infarction responses. BMC Genomics 2015; 16 Suppl 7:S18. [PMID: 26100218 PMCID: PMC4474415 DOI: 10.1186/1471-2164-16-s7-s18] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Pathway analysis has been widely used to gain insight into essential mechanisms of the response to myocardial infarction (MI). Currently, there exist multiple pathway databases that organize molecular datasets and manually curate pathway maps for biological interpretation at varying forms of organization. However, inconsistencies among different databases in pathway descriptions, frequently due to conflicting results in the literature, can generate incorrect interpretations. Furthermore, although pathway analysis software provides detailed images of interactions among molecules, it does not exhibit how pathways interact with one another or with other biological processes under specific conditions. Methods We propose a novel method to standardize descriptions of enriched pathways for a set of genes/proteins using Gene Ontology terms. We used this method to examine the relationships among pathways and biological processes for a set of condition-specific genes/proteins, represented as a functional biological pathway-process network. We applied this algorithm to a set of 613 MI-specific proteins we previously identified. Results A total of 96 pathways from Biocarta, KEGG, and Reactome, and 448 Gene Ontology Biological Processes were enriched with these 613 proteins. The pathways were represented as Boolean functions of biological processes, delivering an interactive scheme to organize enriched information with an emphasis on involvement of biological processes in pathways. We extracted a network focusing on MI to demonstrate that tyrosine phosphorylation of Signal Transducer and Activator of Transcription (STAT) protein, positive regulation of collagen metabolic process, coagulation, and positive/negative regulation of blood coagulation have immediate impacts on the MI response. Conclusions Our method organized biological processes and pathways in an unbiased approach to provide an intuitive way to identify biological properties of pathways under specific conditions. Pathways from different databases have similar descriptions yet diverse biological processes, indicating variation in their ability to share similar functional characteristics. The coverages of pathways can be expanded with the incorporation of more biological processes, predicting involvement of protein members in pathways. Further, detailed analyses of the functional biological pathway-process network will allow researchers and scientists to explore critical routes in biological systems in the progression of disease.
Collapse
|
9
|
Sridharan V, Tripathi P, Sharma SK, Moros EG, Corry PM, Lieblong BJ, Kaschina E, Unger T, Thöne-Reineke C, Hauer-Jensen M, Boerma M. Cardiac inflammation after local irradiation is influenced by the kallikrein-kinin system. Cancer Res 2012; 72:4984-92. [PMID: 22865451 PMCID: PMC3463770 DOI: 10.1158/0008-5472.can-12-1831] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Radiotherapy of intrathoracic and chest wall tumors may lead to exposure of the heart to ionizing radiation, resulting in radiation-induced heart diseases (RIHD). The main manifestations of RIHD become apparent many years after treatment and include cardiomyopathy and accelerated atherosclerosis. This study examines the role of the kallikrein-kinin system (KKS) in RIHD by investigating the cardiac radiation response in a kininogen-deficient Brown Norway Katholiek (BN/Ka) rat model. BN/Ka rats and wild-type Brown Norway (BN) rats were exposed to local heart irradiation with a single dose of 18 Gy or 24 Gy and were observed for 3 to 6 months. Examinations included in vivo and ex vivo cardiac function, histopathology, gene and protein expression measurements, and mitochondrial swelling assays. Upon local heart irradiation, changes in in vivo cardiac function were significantly less in BN/Ka rats. For instance, a single dose of 24 Gy caused a 35% increase in fractional shortening in BN rats compared with a 16% increase in BN/Ka rats. BN rats, but not BN/Ka rats, showed a 56% reduction in cardiac numbers of CD2-positive cells, and a 57% increase in CD68-positive cells, together with a 52% increase in phosphorylation of extracellular signal-regulated kinase 1/2 (Erk1/2). Local heart irradiation had similar effects on histopathology, mitochondrial changes, and left ventricular mRNA levels of NADPH oxidases in the two genotypes. These results suggest that the KKS plays a role in the effects of radiation on cardiac function and recruitment of inflammatory cells. The KKS may have these effects at least in part by altering Erk1/2 signaling.
Collapse
Affiliation(s)
- Vijayalakshmi Sridharan
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Division of Radiation Health, Little Rock, Arkansas
| | - Preeti Tripathi
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Division of Radiation Health, Little Rock, Arkansas
| | - Sunil K. Sharma
- University of Arkansas for Medical Sciences, Department of Radiation Oncology, Little Rock, Arkansas
| | - Eduardo G. Moros
- Moffitt Cancer Center and Research Institute, Department of Radiation Oncology, Tampa, Florida
| | - Peter M. Corry
- University of Arkansas for Medical Sciences, Department of Radiation Oncology, Little Rock, Arkansas
| | - Benjamin J. Lieblong
- University of Arkansas for Medical Sciences, Department of Pharmacology and Toxicology, Little Rock, Arkansas
| | - Elena Kaschina
- Charité University, Institute of Pharmacology, Berlin, Germany
| | - Thomas Unger
- Charité University, Institute of Pharmacology, Berlin, Germany
| | | | - Martin Hauer-Jensen
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Division of Radiation Health, Little Rock, Arkansas
- Surgical Service, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| | - Marjan Boerma
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Division of Radiation Health, Little Rock, Arkansas
| |
Collapse
|
10
|
|
11
|
Duchene J, Bader M. Bradykinin B2 receptor agonism: a novel therapeutic strategy for myocardial infarction? Am J Hypertens 2010; 23:459. [PMID: 20404805 DOI: 10.1038/ajh.2010.32] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
12
|
Cardioprotective effects of a selective B(2) receptor agonist of bradykinin post-acute myocardial infarct. Am J Hypertens 2010; 23:562-8. [PMID: 20186129 DOI: 10.1038/ajh.2010.20] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The cardioprotective benefits of bradykinin are attributable to activation of its B(2) receptor (B(2)R)-mediated actions and abolished by B(2)R antagonists. The current experiments evaluated the cardioprotective potential of a potent, long-acting B(2)R-selective agonist peptide analogue of bradykinin, the compound NG291. METHODS We compared the extent of cardiac tissue damage and remodeling and expression pattern of selected genes in mice submitted to acute myocardial infarct (MI) and treated for 1 week with either NG291 [Hyp(3),Thi(5),(N)Chg(7),Thi(8)]-bradykinin or with saline delivered via osmotic minipump. RESULTS Active treatment resulted in better ejection fraction (EF) 69 +/- 1% vs. 61 +/- 3.1% (P = 0.01), (vs. 85 +/- 1.3% in sham-operated controls), fractional shortening (FS) 38 +/- 4% vs. 32 +/- 8% (NS) (vs. 53 +/- 1.2 in sham-operated controls), and fewer markers of myocyte apoptosis (TUNEL-positive nuclei 4.9 +/- 1.1% vs. 9.7 +/- 0.03%, P = 0.03). Systolic blood pressure (SBP) at end point was normal at 110 +/- 4.2 in actively treated mice, but tended to be lower at 104 +/- 4.7 mm Hg in saline controls with decreased cardiac systolic capacity. Expression patterns of selected genes to factors related to tissue injury, inflammation, and metabolism (i.e., the B(1)R, B(2)R, endothelial nitric oxide synthase (eNOS), TNF-alpha, cardiomyopathy-associated 3 (Cmya3), and pyruvate dehydrogenase kinase isoenzyme 4 (PDK4)) showed that acute MI induced significant upregulation of these genes, and active treatment prevented or attenuated this upregulation, whereas the B(2)R agonist itself produced no difference in the myocardium of sham-operated mice. CONCLUSIONS Treatment with a selective B(2)R agonist initiated at the time of induction of acute MI in mice had a beneficial effect on cardiac function, tissue remodeling, and inflammation-related tissue gene expression, which may explain its structural and functional benefits.
Collapse
|
13
|
Katori M, Majima M. A Novel Category of Anti-Hypertensive Drugs for Treating Salt-Sensitive Hypertension on the Basis of a New Development Concept. Pharmaceuticals (Basel) 2010; 3:59-109. [PMID: 27713243 PMCID: PMC3991021 DOI: 10.3390/ph3010059] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2009] [Revised: 12/24/2009] [Accepted: 01/06/2010] [Indexed: 12/20/2022] Open
Abstract
Terrestrial animals must conserve water and NaCl to survive dry environments. The kidney reabsorbs 95% of the sodium filtered from the glomeruli before sodium reaches the distal connecting tubules. Excess sodium intake requires the renal kallikrein-kinin system for additional excretion. Renal kallikrein is secreted from the distal connecting tubule cells of the kidney, and its substrates, low molecular kininogen, from the principal cells of the cortical collecting ducts (CD). Formed kinins inhibit reabsorption of NaCl through bradykinin (BK)-B₂ receptors, localized along the CD. Degradation pathway of BK by kinin-destroying enzymes in urine differs completely from that in plasma, so that ACE inhibitors are ineffective. Urinary BK is destroyed mainly by a carboxypeptidase-Y-like exopeptidase (CPY) and partly by a neutral endopeptidase (NEP). Inhibitors of CPY and NEP, ebelactone B and poststatin, respectively, were found. Renal kallikrein secretion is accelerated by potassium and ATP-sensitive potassium (KATP) channel blockers, such as PNU-37883A. Ebelactone B prevents DOCA-salt hypertension in rats. Only high salt intake causes hypertension in animals deficient in BK-B2 receptors, tissue kallikrein, or kininogen. Hypertensive patients, and spontaneously hypertensive rats, excrete less kallikrein than normal subjects, irrespective of races, and become salt-sensitive. Ebelactone B, poststatin, and KATP channel blockers could become novel antihypertensive drugs by increase in urinary kinin levels. Roles of kinin in cardiovascular diseases were discussed.
Collapse
Affiliation(s)
- Makoto Katori
- Department of Pharmacology, School of Medicine, Kitasato University, Sagamihara, Kanagawa 228-8555, Japan.
| | - Masataka Majima
- Department of Pharmacology, School of Medicine, Kitasato University, Sagamihara, Kanagawa 228-8555, Japan
| |
Collapse
|
14
|
Tang M, Cui M, Dong Q, Ren HM, Xiao BG, Luo BY, Shao Y, Liu L, Zhou HG. The bradykinin B2 receptor mediates hypoxia/reoxygenation induced neuronal cell apoptosis through the ERK1/2 pathway. Neurosci Lett 2009; 450:40-4. [DOI: 10.1016/j.neulet.2008.10.110] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2008] [Revised: 10/19/2008] [Accepted: 10/31/2008] [Indexed: 11/30/2022]
|
15
|
Dabek J, Kulach A, Smolka G, Wilczok T, Scieszka J, Gasior Z. Expression of genes encoding kinin receptors in peripheral blood mononuclear cells from patients with acute coronary syndromes. Intern Med J 2008; 38:892-6. [DOI: 10.1111/j.1445-5994.2008.01718.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
16
|
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]
|
17
|
Valenti C, Cialdai C, Giuliani S, Tramontana M, Quartara L, Maggi CA. MEN16132, a kinin B2 receptor antagonist, prevents the endogenous bradykinin effects in guinea-pig airways. Eur J Pharmacol 2007; 579:350-6. [PMID: 18036587 DOI: 10.1016/j.ejphar.2007.10.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2007] [Revised: 10/12/2007] [Accepted: 10/18/2007] [Indexed: 10/22/2022]
Abstract
Kinins have been suggested to be involved in human airway diseases such as asthma and rhinitis. MEN16132 is a non-peptide kinin B(2) receptor antagonist able to inhibit the responses produced by intravenous bradykinin into the airways, as bronchoconstriction and microvascular leakage; we tested the effect of MEN16132 on endogenously generated bradykinin through the dextran sulfate-induced contact activation of kinin-kallikrein cascade in guinea-pigs. After dextran sulfate administration (1.5 mg/kg i.v.), the pulmonary insufflation pressure was monitored and the microvascular leakage of upper and lower airways was assessed using Evans blue as tracer of plasma protein extravasation. Our results demonstrated that topical MEN16132 strongly inhibited the dextran sulfate-induced bronchoconstriction (0.3 mM solution aerosol for 5 min) and plasma protein extravasation in both lower airways (3-10 microM solution aerosol for 5 min) and nasal mucosa (0.3 nmol/nostril); Icatibant, the peptide antagonist of kinin B(2) receptor, exerted a 3-30-fold less potent inhibitory effect than MEN16132. We conclude that local application of MEN16132 into the airways abolishes the responses produced by the endogenous generation of bradykinin and it can be useful as new pharmacological tool to check the role of kinins in human diseases.
Collapse
Affiliation(s)
- Claudio Valenti
- Pharmacology Department, Menarini Ricerche S.pA.,Via Rismondo 12A, 50131 Florence, Italy
| | | | | | | | | | | |
Collapse
|
18
|
Yin H, Chao J, Bader M, Chao L. Differential role of kinin B1 and B2 receptors in ischemia-induced apoptosis and ventricular remodeling. Peptides 2007; 28:1383-9. [PMID: 17644219 PMCID: PMC2067250 DOI: 10.1016/j.peptides.2007.05.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Revised: 05/15/2007] [Accepted: 05/16/2007] [Indexed: 11/17/2022]
Abstract
We investigated the role of kinin receptors in cardiac remodeling after ischemia/reperfusion (I/R). Bradykinin injection improved cardiac contractility, diastolic function, reduced infarct size and prevented left ventricular thinning after I/R, whereas des-Arg(9)-BK injection had no protective effects. Bradykinin, but not des-Arg(9)-BK, reduced cardiomyocyte apoptosis and increased Akt and GSK-3beta phosphorylation. Furthermore, myocardial infarct size was similar between wild type and B2 knockout mice after I/R, but significantly reduced in kinin B1 receptor knockout mice. These results indicate that the kinin B2 receptor, but not the B1 receptor, protects against I/R-induced cardiac dysfunction by inhibiting apoptosis and limiting ventricular remodeling.
Collapse
Affiliation(s)
- Hang Yin
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425-2211, USA
| | | | | | | |
Collapse
|
19
|
Abstract
The myocardium represents a major source of several families of peptide hormones under normal physiological conditions and the plasma concentrations of many of these "cardiac peptides" (or related pro-peptide fragments) are substantially augmented in many cardiac disease states. In addition to well-characterised endocrine functions of several of the cardiac peptides, pleiotropic functions within the myocardium and the coronary vasculature represent a significant aspect of their actions in health and disease. Here, we focus specifically on the cardioprotective roles of four major peptide families in myocardial ischemia and reperfusion: adrenomedullin, kinins, natriuretic peptides and the urocortins. The patterns of early release of all these peptides are consistent with roles as autacoid cardioprotective mediators. Clinical and experimental research indicates the early release and upregulation of many of these peptides by acute ischemia and there is a convincing body of evidence showing that exogenously administered adrenomedullin, bradykinin, ANP, BNP, CNP and urocortins are all markedly protective against experimental myocardial ischemia-reperfusion injury through a conserved series of cytoprotective signal transduction pathways. Intriguingly, all the peptides examined so far have the potential to salvage against infarction when administered specifically during early reperfusion. Thus, the myocardial secretion of peptide hormones likely represents an early protective response to ischemia. Further work is required to explore the potential therapeutic manipulation of these peptides in acute coronary syndromes and their promise as biomarkers of acute myocardial ischemia.
Collapse
Affiliation(s)
- Dwaine S Burley
- Department of Basic Sciences, The Royal Veterinary College, University of London, Royal College Street, London, UK
| | | | | |
Collapse
|
20
|
Fukushima S, Coppen SR, Varela-Carver A, Yamahara K, Sarathchandra P, Smolenski RT, Yacoub MH, Suzuki K. A novel strategy for myocardial protection by combined antibody therapy inhibiting both P-selectin and intercellular adhesion molecule-1 via retrograde intracoronary route. Circulation 2006; 114:I251-6. [PMID: 16820581 DOI: 10.1161/circulationaha.105.000794] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Antibody therapy to inhibit either P-selectin or intercellular adhesion molecule-1 (ICAM-1) has been reported to provide myocardial protection against leukocyte-mediated reperfusion injury. Because these molecules play different roles in the leukocyte-endothelial interaction, co-inhibition of both may achieve further enhanced cardioprotection. In addition, the therapeutic efficacy of such antibody therapy may be affected by the delivery route used. Retrograde intracoronary infusion will offer an effective, direct access to the postcapillary venules, where the target event (leukocyte-endothelial interaction) takes place. We investigated the feasibility and efficiency of the combined antibody therapy targeting both P-selection and ICAM-1 via the retrograde intracoronary route to attenuate myocardial ischemia-reperfusion injury. METHODS AND RESULTS Lewis rats underwent 30-minute left coronary artery occlusion. Just before reperfusion, anti-P-selectin monoclonal antibody (150 microg/kg), anti-ICAM-1 monoclonal antibody (200 microg/kg), both antibodies together, or control antibody were retrogradely infused into the left cardiac vein. At 24 hours after reperfusion, administration of either anti-P-selectin or anti-ICAM-1 antibody significantly (P<0.05) improved left ventricular ejection fraction and attenuated infarct size (40.6+/-3.2% and 34.8+/-3.5%, respectively) compared with the control (56.8+/-3.4%). This was associated with reduced leukocyte accumulation and improved regional blood flow in the ischemic area. Noticeably, co-administration of both antibodies achieved a much greater reduction in infarct size (19.1+/-3.6%), associated with greater attenuation in leukocyte infiltration, compared with administration of either single antibody. CONCLUSIONS Combined antibody therapy inhibiting both P-selectin and ICAM-1 via the retrograde intracoronary route could be a promising new strategy for myocardial protection against ischemia-reperfusion injury.
Collapse
MESH Headings
- Acute Disease
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Chemotaxis, Leukocyte/drug effects
- Coronary Circulation
- Coronary Vessels
- Drug Evaluation, Preclinical
- Drug Synergism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/immunology
- Feasibility Studies
- Heart Ventricles/diagnostic imaging
- Injections, Intravenous/methods
- Intercellular Adhesion Molecule-1/immunology
- Leukocytes/drug effects
- Leukocytes/immunology
- Male
- Myocardial Infarction/complications
- Myocardial Infarction/pathology
- Myocardial Reperfusion Injury/prevention & control
- Myocarditis/etiology
- Myocarditis/pathology
- Myocarditis/prevention & control
- Organ Size
- P-Selectin/immunology
- Rats
- Rats, Inbred Lew
- Stroke Volume
- Ultrasonography
Collapse
Affiliation(s)
- Satsuki Fukushima
- Harefield Heart Science Centre, National Heart & Lung Institute, Faculty of Medicine, Imperial College London, UK
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Koike MK, de Carvalho Frimm C, de Lourdes Higuchi M. Bradykinin B2receptor antagonism attenuates inflammation, mast cell infiltration and fibrosis in remote myocardium after infarction in rats. Clin Exp Pharmacol Physiol 2006; 32:1131-6. [PMID: 16445581 DOI: 10.1111/j.1440-1681.2005.04309.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Bradykinin may interfere with myocardial remodelling by promoting inflammation and mast cell activation or, alternatively, by counteracting angiotensin II-dependent collagen accumulation. The aim of the present study was to investigate the role of bradykinin B2 receptor antagonism in inflammatory and mast cell infiltration, fibroplasia and fibrosis accumulation following myocardial infarction (MI). Myocardial infarction was produced by the ligature of the left coronary artery in male Wistar rats that were 10 weeks of age. Immediately after MI, rats received the B2 receptor antagonist Hoe140 (0.5 microg/kg per min, s.c.) or saline over a period of 3 days, 1 week or 4 weeks, constituting three separate groups and their respective controls. Coronal myocardial tissue sections underwent haematoxylin and eosin, Giemsa and picrosirius red staining, as well as immunohistochemistry for alpha-smooth muscle actin (SMA). Morphometric studies were undertaken in three different myocardial regions: MI, remote non-infarcted subendocardium (non-MI SE) and remote non-infarcted interventricular septum (non-MI IVS). The MI size was comparable between Hoe140-treated groups and their respective controls (day 3: 42 +/- 4%, n = 8, vs 43 +/- 3%, n = 6; week 1: 37 +/- 5%, n = 5, vs 39 +/- 2%, n = 5; week 4: 35 +/- 3%, n = 9, vs 36 +/- 3%, n = 7). At day 3, Hoe140 treatment reduced inflammatory cell reaction within the MI (585 +/- 59 vs 995 +/- 170 cells/mm2; P = 0.02), non-MI SE (77 +/- 12 vs 214 +/- 57 cells/mm2; P = 0.02) and non-MI IVS (93 +/- 16 vs 135 +/- 14 cells/mm2; P = 0.03) regions. Mast cells were reduced within the non-MI IVS region (0.8 +/- 0.1 vs 2.5 +/- 0.4 cells/mm2; P = 0.006), but not within the MI region. In non-MI SE, mast cells were rarely found. At week 1, Hoe140 treatment reduced alpha-SMA-positive myofibroblast infiltration within the MI (2535 +/- 383 vs 5636 +/- 968 cells/mm2; P = 0.01) and non-MI SE (222 +/- 33 vs 597 +/- 162 cells/mm2; P = 0.03) regions. In the non-MI IVS region, alpha-SMA-positive myofibroblasts were rarely found. At week 4, Hoe140 treatment reduced collagen volume fraction within the MI (37 +/- 4 vs 53 +/- 4%; P = 0.03), non-MI SE (1.3 +/- 0.2 vs 2.6 +/- 0.3%; P = 0.001) and non-MI IVS (1.1 +/- 0.2 vs 1.8 +/- 0.2%; P = 0.01) regions. Bradykinin promotes inflammation, fibroplasia and fibrosis after MI. Mast cells may have a role in fibrosis deposition through a bradykinin-related mechanism.
Collapse
Affiliation(s)
- Marcia Kiyomi Koike
- LIM-51, Emergências Clínicas, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | | |
Collapse
|
22
|
Valenti C, Cialdai C, Giuliani S, Lecci A, Tramontana M, Meini S, Quartara L, Maggi CA. MEN16132, a novel potent and selective nonpeptide kinin B2 receptor antagonist: in vivo activity on bradykinin-induced bronchoconstriction and nasal mucosa microvascular leakage in anesthetized guinea pigs. J Pharmacol Exp Ther 2005; 315:616-23. [PMID: 16027229 DOI: 10.1124/jpet.105.088252] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have tested the activity of 4-(S)-amino-5-(4-[4-[2,4-dichloro-3-(2,4-dimethyl-8-quinolyloxymethyl)phenylsulfonamido]-tetrahydro-2H-4-pyranylcarbonyl] piperazino)-5-oxopentyl](trimethyl)ammonium chloride hydrochloride (MEN16132), a novel nonpeptide kinin B(2) receptor antagonist, on bradykinin (BK)-induced inflammatory responses, bronchoconstriction, and hypotension in guinea pigs. After i.v. (1-10 nmol/kg i.v.), intratracheal (i.t.) (10-100 nmol/kg i.t.), or aerosol (0.01-0.1 mM/5 min) administration, MEN16132 inhibited in a dose-dependent manner the bronchoconstriction induced by BK (10 nmol/kg i.v.). MEN16132 was more potent and possessed a longer duration of action as compared with the peptide B(2) receptor antagonist icatibant (HOE140; H-D-Arg-Arg-Pro-Hyp-Gly-Thi-Ser-D-Tic-Oic-Arg-OH trifluoroacetate). After i.v. administration, its inhibitory effect on bronchoconstriction lasted more than 8 h at 30 nmol/kg. When administered by i.v. or i.t. routes, the dose completely inhibiting bronchoconstriction also partially reduced the hypotensive response to BK, whereas after aerosol administration, the inhibitory effect was limited to respiratory level. Intranasal (i.n.) administration of MEN16132 (0.01-0.3 nmol/nostril) reduced, in a dose-dependent and long-lasting manner, the nasal mucosa plasma protein extravasation induced by BK (100 nmol/nostril), and it exerted a complete inhibition at about 30-fold lower dose than icatibant. At 1 nmol/nostril, MEN16132 activity was significant for at least 6 h with no systemic effect measured as inhibition of BK-induced hypotension, and at 10 nmol/nostril, the inhibitory effect lasted for more than 15 h with only a weak effect on hypotension. These findings indicate that in vivo MEN16132 is a potent kinin B(2) receptor antagonist with long duration of action, both after i.v. and local administration. A complete and prolonged inhibition of BK-induced bronchoconstriction or nasal inflammation can be achieved with MEN16132 topical administration (aerosol or i.n.) at doses devoid of systemic effects.
Collapse
Affiliation(s)
- Claudio Valenti
- Departments of Pharmacology, Menarini Ricerche, Florence, Italy
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Nomura I, Abe J, Noma S, Saito H, Gao B, Wheeler G, Leung DYM. Adrenomedullin is highly expressed in blood monocytes associated with acute Kawasaki disease: a microarray gene expression study. Pediatr Res 2005; 57:49-55. [PMID: 15531734 DOI: 10.1203/01.pdr.0000147745.52711.dd] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Kawasaki disease (KD) is an acute inflammatory disorder of children frequently associated with the development of coronary artery abnormalities. Although a great deal is known about inflammatory and immune responses in acute KD, the mechanisms linking the immune response to vascular changes are not known. To gain further insight into this process, we performed a microarray gene expression analysis on RNA isolated from the peripheral blood mononuclear cells of four patients with KD during both their acute and convalescent phases. Forty-seven genes of 7129 genes examined showed an increased expression in three or all four patients in the acute compared with the convalescent phase of KD. Fourteen of these genes were significantly (p < 0.05) up-regulated, including several inflammatory response genes (e.g. S-100 A9 protein) and also anti-inflammatory genes (e.g. TSG-6). Of greatest interest, the adrenomedullin (ADM) gene, known to be associated with coronary artery vasodilation, was up-regulated in the acute phase of KD (p = 0.024). Up-regulation of ADM in the acute phase of KD was confirmed in peripheral blood mononuclear cells of 11 additional KD patients by reverse transcriptase-PCR (p < 0.01). Isolated blood monocytes but not lymphocytes were demonstrated by real-time PCR to have increased ADM mRNA (p = 0.01). Plasma ADM protein level in 32 additional KD patients was also confirmed to be higher in acute KD compared with convalescent KD (p < 0.032). It is interesting that from microarray results, other molecules known to be associated with coronary dilation, including nitric oxide, prostacyclin, acetylcholine, bradykinin, substance P, and serotonin, were not elevated in acute KD. Our current study suggests that ADM-expressing monocytes that infiltrate the coronary vascular wall may be the cause of coronary dilation in the acute phase of KD.
Collapse
Affiliation(s)
- Ichiro Nomura
- Department of Pediatrics, National Jewish Medical and Research Center, Denver, CO 80206, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Fischer M, Lieb W, Marold D, Berthold M, Baessler A, Lowel H, Hense HW, Hengstenberg C, Holmer S, Schunkert H, Erdmann J. Lack of association of a 9 bp insertion/deletion polymorphism within the bradykinin 2 receptor gene with myocardial infarction. Clin Sci (Lond) 2004; 107:505-11. [PMID: 15301669 DOI: 10.1042/cs20040129] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The BK (bradykinin) B2 receptor is the major cellular mediator of the effects of BK. A 9 bp deletion in the promoter of the receptor gene represents an allelic variant that is associated with enhanced mRNA expression levels. We tested whether this polymorphism is associated with the prevalence of MI (myocardial infarction) or with echocardiographically determined left ventricular function in post-MI patients. Patients with documented MI (n=484), matched controls and controls without evidence of coronary heart disease (n=1363) constituted cases and controls. MI patients and controls were carefully matched for age, gender and cardiovascular risk factors. Genotype distributions of the 9 bp insertion/deletion polymorphism were similar across the groups: −9/−9, −9/+9 and +9/+9 were 22.1, 49.5 and 28.5% in MI patients, and 23.0, 44.6 and 32.5% in matched control subjects respectively. The lack of association was also observed in selected subgroups, stratified by age, gender and cardiovascular risk factors. Furthermore, there was no relation between this polymorphism and left ventricular systolic function in post-MI patients. These findings indicate that the 9 bp insertion/deletion polymorphism of the BK B2 receptor gene is neither related to the prevalence of MI nor to left ventricular function after MI.
Collapse
Affiliation(s)
- Marcus Fischer
- Clinic for Internal Medicine 2, University of Regensburg, Franz-Josef-Strauss Allee 11, 93053 Regensburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bellucci F, Meini S, Cucchi P, Catalani C, Reichert W, Zappitelli S, Rotondaro L, Quartara L, Giolitti A, Maggi CA. A different molecular interaction of bradykinin and the synthetic agonist FR190997 with the human B2 receptor: evidence from mutational analysis. Br J Pharmacol 2003; 140:500-6. [PMID: 12970081 PMCID: PMC1574048 DOI: 10.1038/sj.bjp.0705454] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Binding affinity at the [3H]-BK binding site and activity as inositol phosphate (IP) production by the peptide bradykinin (BK) and the nonpeptide FR190997 were studied at wild-type or point-mutated human B2 receptors (hB2R) expressed in CHO cells. The effect of the following mutations were analyzed: E47A (TM1), W86A and T89A (TM2), I110A, L114A and S117A (TM3), T158A, M165T and L166F (TM4), T197A and S211A (TM5), F252A, W256A and F259A (TM6), S291A, F292A, Y295A and Y295F (TM7), and the double mutation W256A/Y295F. As the wild-type receptor-binding affinity of FR190997 was 40-fold lower than BK, whereas their agonist potency was comparable, both agonists produced similar maximal effects (Emax). Mutations were evaluated as affecting the affinity and/or efficacy of FR190997 compared with BK. Two mutations were found to impair the agonist affinity of both agonists drastically: W86A and F259A. BK agonist affinity (pEC50) was reduced by 1400- and 150-fold, and that of FR190997 was reduced by 400- and 25-fold, at the W86A and F259A mutant B2 receptors, respectively. Contrary to BK, the affinity of FR190997 was selectively decreased at I110A, Y295A, and Y295F mutants (>103-fold), and a different efficacy was measured at the Y295 mutants, FR190997 being devoid of the capability to trigger IP production at Y295A mutant. L114A, F252A, and W256A selectively impaired the efficacy of FR190997, whereas its binding affinity was not affected. As a consequence, FR190997 behaved as a high-affinity antagonist in blocking the IP production induced by BK. The lack of capability of FR190997 to activate or to bind the double mutant W256A/Y295F suggests that these residues are part of the same binding site, which is also important for receptor activation by the nonpeptide ligand. Overall, by means of mutational analysis, we indicate an hB2R recognition site for the nonpeptide agonist FR190997 (between TM3, 6, and 7), different from that of BK, and show that in the same binding crevice some mutations (L114, W256, and F252) are selectively responsible for the agonist properties of only FR190997.
Collapse
Affiliation(s)
- Francesca Bellucci
- Department of Pharmacology, Menarini Ricerche S.p.A., via Rismondo 12A, Florence 50131, Italy
| | - Stefania Meini
- Department of Pharmacology, Menarini Ricerche S.p.A., via Rismondo 12A, Florence 50131, Italy
- Author for correspondence:
| | - Paola Cucchi
- Department of Pharmacology, Menarini Ricerche S.p.A., via Rismondo 12A, Florence 50131, Italy
| | - Claudio Catalani
- Department of Pharmacology, Menarini Ricerche S.p.A., via Rismondo 12A, Florence 50131, Italy
| | | | - Sabrina Zappitelli
- Department of Biotechnology, Menarini Biotech, via Tito Speri 10, Rome 00040, Italy
| | - Luigi Rotondaro
- Department of Biotechnology, Menarini Biotech, via Tito Speri 10, Rome 00040, Italy
| | - Laura Quartara
- Department of Chemistry, Menarini Ricerche S.p.A., via Rismondo 12A, Florence 50131, Italy
| | - Alessandro Giolitti
- Department of Drug Design, Menarini Ricerche S.p.A., via Rismondo 12A, Florence 50131, Italy
| | - Carlo Alberto Maggi
- Department of Pharmacology, Menarini Ricerche S.p.A., via Rismondo 12A, Florence 50131, Italy
| |
Collapse
|