1
|
El Cheikh J, Hamed F, Rifi H, Dakroub AH, Eid AH. Genetic polymorphisms influencing antihypertensive drug responses. Br J Pharmacol 2025; 182:929-950. [PMID: 39627167 DOI: 10.1111/bph.17414] [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: 05/21/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 01/11/2025] Open
Abstract
Hypertension is a major contributor to cardiovascular disease and its associated morbidity and mortality. The low efficacy observed with some anti-hypertensive therapies has been attributed partly to inter-individual genetic variability. This paper reviews the major findings regarding these genetic variabilities that modulate responses to anti-hypertensive therapies such as angiotensin converting enzyme (ACE) inhibitors, angiotensin receptor blockers (ARBs), diuretics, calcium channel blockers (CCBs) and β-adrenoceptor blockers. The importance of studying these genetic polymorphisms stems from the goal to optimise anti-hypertensive therapy for each individual patient, aiming for the highest efficacy and lowest risk of adverse effects. It is important to recognise that environmental and epigenetic factors can contribute to the observed variations in drug responses. Owing to the multigenic and multifactorial nature of drug responses, further research is crucial for translating these findings into clinical practice and the establishment of reliable recommendations.
Collapse
Affiliation(s)
- Jana El Cheikh
- Faculty of Medicine, University of Balamand, Al Koura, Tripoli, Lebanon
| | - Fouad Hamed
- Faculty of Medicine, University of Balamand, Al Koura, Tripoli, Lebanon
| | - Hana Rifi
- Faculty of Medicine, University of Balamand, Al Koura, Tripoli, Lebanon
| | - Ali H Dakroub
- Blavatnik Family Research Institute, Departments of Cardiology and Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ali Hussein Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
2
|
Rodríguez JE, Andrade-Jorge E, Barquet-Nieto A, Estrada-Soto SE, Gallardo-Ortíz IA, Villalobos-Molina R. BMY 7378, a selective α 1D-adrenoceptor antagonist, is a new angiotensin converting enzyme inhibitor: In silico, in vitro and in vivo approach. Biochim Biophys Acta Gen Subj 2025; 1869:130732. [PMID: 39631474 DOI: 10.1016/j.bbagen.2024.130732] [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: 07/17/2024] [Revised: 11/11/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
BMY 7378 is a multitarget drug primarily known for its selective antagonism of α1D-adrenoceptors (α1D-AR), exhibiting both hypotensive effects and the ability to prevent or reverse angiotensin II-induced vascular hypertrophy. Notably, BMY 7378 contains a phenylpiperazine moiety, a structural feature associated with angiotensin-converting enzyme (ACE) inhibition. This study aimed to investigate ACE inhibition as a potential pharmacological mechanism of BMY 7378. Using an in silico approach we predicted BMY 7378 interactions with the ACE active site, followed by in vitro activity assays. Additionally, ACE protein expression in the heart was analyzed following four weeks of BMY 7378 treatment in 7-8-month-old spontaneously hypertensive rats (SHR). All assays were benchmarked against captopril, a standard ACE inhibitor. In silico results showed that BMY 7378 binds to the ACE active site, though with reduced interaction with Zn701 (73.7 % compared to captopril), likely due to the pKa of its amino group. The inhibitory concentration 50 (IC50) for BMY 7378 was 136 μM, lower than other reported phenylpiperazine derivatives. Furthermore, BMY 7378 significantly increased ACE expression in the hearts of SHR, with an increase of 8.5-fold compared to captopril. In conclusion, BMY 7378 exhibits dual activity as an α1D-AR antagonist and an ACE inhibitor, making it a promising pharmacological tool for investigating and potentially treating hypertension and its associated cardiovascular complications.
Collapse
Affiliation(s)
- Jessica E Rodríguez
- Bioquímica Clínica, Carrera de Químico Farmacéutico Biólogo, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Av. Guelatao con Av. Exploradores, Ejército de Oriente, Iztapalapa, 09230, Ciudad de Mexico, México; Unidad de Investigación en Biomedicina, Carrera de Enfermería, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Estado de México, México
| | - Erik Andrade-Jorge
- Laboratorio de Investigación en Bioquímica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n. Casco de Santo Tomás, 11340 Ciudad de México, México
| | - Alina Barquet-Nieto
- Laboratorio de Investigación en Bioquímica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n. Casco de Santo Tomás, 11340 Ciudad de México, México
| | - Samuel E Estrada-Soto
- Laboratorio de Farmacognosia, Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Avenida Universidad 1001, Chamilpa, 62209 Cuernavaca, Morelos, México
| | - Itzell A Gallardo-Ortíz
- Unidad de Investigación en Biomedicina, Carrera de Enfermería, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Estado de México, México.
| | - Rafael Villalobos-Molina
- Unidad de Investigación en Biomedicina, Carrera de Enfermería, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Estado de México, México.
| |
Collapse
|
3
|
Kopaliani I, Elsaid B, Speier S, Deussen A. Immune and Metabolic Mechanisms of Endothelial Dysfunction. Int J Mol Sci 2024; 25:13337. [PMID: 39769104 PMCID: PMC11728141 DOI: 10.3390/ijms252413337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/01/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
Endothelial dysfunction is a strong prognostic factor in predicting the development of cardiovascular diseases. Dysfunctional endothelium loses its homeostatic ability to regulate vascular tone and prevent overactivation of inflammation, leading to vascular dysfunction. These functions are critical for vascular homeostasis and arterial pressure control, the disruption of which may lead to hypertension. Hypertension itself can also cause endothelial dysfunction, as endothelial cells are susceptible to haemodynamic changes. Although it is unclear which of those factors appear first, they create a vicious circle further damaging multiple organs, including the heart and vessels. There are also sex-specific differences in homeostatic functions of the endothelium regarding vessel tone regulation, which may contribute to differences in arterial blood pressure between men and women. Even more importantly, there are sex-differences in the development of endothelial dysfunction and vessel remodelling. Hence, an understanding of the mechanisms of endothelial dysfunction and its contribution to pathological vascular remodelling during hypertension is of critical importance. This review addresses immunological and metabolic aspects in mechanisms of endothelial dysfunction and the resulting mechanisms in vascular remodelling with respect to arterial hypertension, including the potential role of sex-specific differences.
Collapse
Affiliation(s)
- Irakli Kopaliani
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
| | - Basant Elsaid
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
- Department of Physiology, Faculty of Medicine, Ain Shams University, Cairo 1181, Egypt
| | - Stephan Speier
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Andreas Deussen
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
| |
Collapse
|
4
|
Bhilare KD, Dobariya P, Hanak F, Rothwell PE, More SS. Current understanding of the link between angiotensin-converting enzyme and pain perception. Drug Discov Today 2024; 29:104089. [PMID: 38977123 PMCID: PMC11368640 DOI: 10.1016/j.drudis.2024.104089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/10/2024]
Abstract
The renin-angiotensin system (RAS) is known to affect diverse physiological processes that affect the functioning of many key organs. Angiotensin-converting enzyme (ACE) modulates a variety of bioactive peptides associated with pain. ACE inhibitors (ACEis) have found applications in the treatment of cardiovascular, kidney, neurological and metabolic disorders. However, ACEis also tend to display undesirable effects, resulting in increased pain sensitization and mechanical allodynia. In this review, we provide comprehensive discussion of preclinical and clinical studies involving the evaluation of various clinically approved ACEis. With the emerging knowledge of additional factors involved in RAS signaling and the indistinct pharmacological role of ACE substrates in pain, extensive studies are still required to elucidate the mechanistic role of ACE in pain perception.
Collapse
Affiliation(s)
- Kiran D Bhilare
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Prakashkumar Dobariya
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Filip Hanak
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Patrick E Rothwell
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Swati S More
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
5
|
Mohammadi K, Shafie D, Ghomashi N, Abdolizadeh A, Sadeghpour M. Kinin-kallikrein system: New perspectives in heart failure. Heart Fail Rev 2024; 29:729-737. [PMID: 38381277 DOI: 10.1007/s10741-024-10393-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 02/22/2024]
Abstract
Heart failure (HF) is a pervasive clinical challenge characterized by compromised cardiac function and reduced quality of life. The kinin-kallikrein system (KSS), a multifaceted peptide cascade, has garnered substantial attention due to its potential role in HF. Through activation of B1 and/or B2 receptors and downstream signaling, kinins modulate various physiological processes, including inflammation, coagulation, pain, blood pressure control, and vascular permeability. Notably, aberrations in KKS components have been linked to HF risk. The elevation of vasodilatory bradykinin (BK) due to kallikrein activity reduces preload and afterload, while concurrently fostering sodium reabsorption inhibition. However, kallikrein's conversion of prorenin to renin leads to angiotensinsII upregulation, resulting in vasoconstriction and fluid retention, alongside increased immune cell activity that fuels inflammation and cardiac remodeling. Importantly, prolonged KKS activation resulting from volume overload and tissue stretch contributes to cardiac collagen loss. The conventional renin-angiotensin-aldosterone system (RAAS) inhibitors used in HF management may inadvertently intensify KKS activity, exacerbating collagen depletion and cardiac remodeling. It is crucial to balance the KKS's role in acute cardiac damage, which may temporarily enhance function and metabolic parameters against its detrimental long-term effects. Thus, KKS blockade emerges as a promising strategy to impede HF progression. By attenuating the link between immune system function and tissue damage, KKS inhibition can potentially reduce cardiac remodeling and alleviate HF symptoms. However, the nuanced roles of BK in various acute conditions necessitate further investigation into the sustained benefits of kallikrein inhibitors in patients with chronic HF.
Collapse
Affiliation(s)
- Keivan Mohammadi
- Shahid Chamran Heart Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Davood Shafie
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Newsha Ghomashi
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Abdolizadeh
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Majid Sadeghpour
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
6
|
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
|
7
|
Li K, Kratzmann V, Dai M, Gatzke N, Rocic P, Bramlage P, Grisk O, Lubomirov LT, Hoffmeister M, Lauxmann MA, Ritter O, Buschmann E, Bader M, Persson AB, Buschmann I, Hillmeister P. Angiotensin receptor-neprilysin inhibitor improves coronary collateral perfusion. Front Cardiovasc Med 2023; 9:981333. [PMID: 36818914 PMCID: PMC9936066 DOI: 10.3389/fcvm.2022.981333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/09/2022] [Indexed: 02/05/2023] Open
Abstract
Background We investigated the pleiotropic effects of an angiotensin receptor-neprilysin inhibitor (ARNi) on collateral-dependent myocardial perfusion in a rat model of coronary arteriogenesis, and performed comprehensive analyses to uncover the underlying molecular mechanisms. Methods A rat model of coronary arteriogenesis was established by implanting an inflatable occluder on the left anterior descending coronary artery followed by a 7-day repetitive occlusion procedure (ROP). Coronary collateral perfusion was measured by using a myocardial particle infusion technique. The putative ARNi-induced pro-arteriogenic effects were further investigated and compared with an angiotensin-converting enzyme inhibitor (ACEi). Expression of the membrane receptors and key enzymes in the natriuretic peptide system (NPS), renin-angiotensin-aldosterone system (RAAS) and kallikrein-kinin system (KKS) were analyzed by quantitative polymerase chain reaction (qPCR) and immunoblot assay, respectively. Protein levels of pro-arteriogenic cytokines were measured by enzyme-linked immunosorbent assay, and mitochondrial DNA copy number was assessed by qPCR due to their roles in arteriogenesis. Furthermore, murine heart endothelial cells (MHEC5-T) were treated with a neprilysin inhibitor (NEPi) alone, or in combination with bradykinin receptor antagonists. MHEC5-T proliferation was analyzed by colorimetric assay. Results The in vivo study showed that ARNis markedly improved coronary collateral perfusion, regulated the gene expression of KKS, and increased the concentrations of relevant pro-arteriogenic cytokines. The in vitro study demonstrated that NEPis significantly promoted MHEC5-T proliferation, which was diminished by bradykinin receptor antagonists. Conclusion ARNis improve coronary collateral perfusion and exert pro-arteriogenic effects via the bradykinin receptor signaling pathway.
Collapse
Affiliation(s)
- Kangbo Li
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Victoria Kratzmann
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Mengjun Dai
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nora Gatzke
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Petra Rocic
- Department of Physiology and Pharmacology, College of Osteopathic Medicine, Sam Houston State University, Huntsville, TX, United States
| | - Peter Bramlage
- Institute for Pharmacology and Preventive Medicine, Cloppenburg, Germany
| | - Olaf Grisk
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Lubomir T. Lubomirov
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Meike Hoffmeister
- Institute of Biochemistry, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg, The Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| | - Martin A. Lauxmann
- Institute of Biochemistry, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Oliver Ritter
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg, The Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
- Department for Cardiology, Center for Internal Medicine I, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Eva Buschmann
- Department of Cardiology, University Clinic Graz, Graz, Austria
| | - Michael Bader
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Center for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Anja Bondke Persson
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ivo Buschmann
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg, The Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| | - Philipp Hillmeister
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg, The Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| |
Collapse
|
8
|
Brusco I, Fialho MFP, Becker G, Brum ES, Favarin A, Marquezin LP, Serafini PT, Oliveira SM. Kinins and their B 1 and B 2 receptors as potential therapeutic targets for pain relief. Life Sci 2023; 314:121302. [PMID: 36535404 DOI: 10.1016/j.lfs.2022.121302] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Kinins are endogenous peptides that belong to the kallikrein-kinin system, which has been extensively studied for over a century. Their essential role in multiple physiological and pathological processes is demonstrated by activating two transmembrane G-protein-coupled receptors, the kinin B1 and B2 receptors. The attention is mainly given to the pathological role of kinins in pain transduction mechanisms. In the past years, a wide range of preclinical studies has amounted to the literature reinforcing the need for an updated review about the participation of kinins and their receptors in pain disorders. Here, we performed an extensive literature search since 2004, describing the historical progress and the current understanding of the kinin receptors' participation and its potential therapeutic in several acute and chronic painful conditions. These include inflammatory (mainly arthritis), neuropathic (caused by different aetiologies, such as cancer, multiple sclerosis, antineoplastic toxicity and diabetes) and nociplastic (mainly fibromyalgia) pain. Moreover, we highlighted the pharmacological actions and possible clinical applications of the kinin B1 and B2 receptor antagonists, kallikrein inhibitors or kallikrein-kinin system signalling pathways-target molecules in these different painful conditions. Notably, recent findings sought to elucidate mechanisms for guiding new and better drug design targeting kinin B1 and B2 receptors to treat a disease diversity. Since the kinin B2 receptor antagonist, Icatibant, is clinically used and well-tolerated by patients with hereditary angioedema gives us hope kinin receptors antagonists could be more robustly tested for a possible clinical application in the treatment of pathological pains, which present limited pharmacology management.
Collapse
Affiliation(s)
- Indiara Brusco
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Maria Fernanda Pessano Fialho
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Gabriela Becker
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Evelyne Silva Brum
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Amanda Favarin
- Laboratory of Neurotoxicity and Psychopharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Lara Panazzolo Marquezin
- Laboratory of Neurotoxicity and Psychopharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Patrick Tuzi Serafini
- Laboratory of Neurotoxicity and Psychopharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Sara Marchesan Oliveira
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
9
|
Abstract
The pineal gland is a interface between light-dark cycle and shows neuro-endocrine functions. Melatonin is the primary hormone of pineal gland, secreted at night. The night-time melatonin peak regulates the physiological functions at dark. Melatonin has several unique features as it synchronises internal rhythm with daily and seasonal variations, regulates circadian rhythm and sleep-wake cycle. Physiologically melatonin involves in detoxification of free radicals, immune functions, neuro-protection, oncostatic effects, cardiovascular functions, reproduction, and foetal development. The precise functions of melatonin are exhibited by specific receptors. In relation to pathophysiology, impaired melatonin secretion promotes sleep disorder, cancer progression, type-2 diabetes, and neurodegenerative diseases. Several reports have highlighted the therapeutic benefits of melatonin specially related to cancer protection, sleep disorder, psychiatric disorders, and jet lag problems. This review will touch the most of the area of melatonin-oriented health impacts and its therapeutic aspects.
Collapse
|
10
|
Alfaro E, Díaz-García E, García-Tovar S, Zamarrón E, Mangas A, Galera R, Nanwani-Nanwani K, Pérez-de-Diego R, López-Collazo E, García-Río F, Cubillos-Zapata C. Impaired Kallikrein-Kinin System in COVID-19 Patients' Severity. Front Immunol 2022; 13:909342. [PMID: 35812405 PMCID: PMC9258198 DOI: 10.3389/fimmu.2022.909342] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/12/2022] [Indexed: 12/27/2022] Open
Abstract
COVID-19 has emerged as a devastating disease in the last 2 years. Many authors appointed to the importance of kallikrein-kinin system (KKS) in COVID-19 pathophysiology as it is involved in inflammation, vascular homeostasis, and coagulation. We aim to study the bradykinin cascade and its involvement in severity of patients with COVID-19. This is an observational cohort study involving 63 consecutive patients with severe COVID-19 pneumonia and 27 healthy subjects as control group. Clinical laboratory findings and plasma protein concentration of KKS peptides [bradykinin (BK), BK1-8], KKS proteins [high–molecular weight kininogen (HK)], and KKS enzymes [carboxypeptidase N subunit 1 (CPN1), kallikrein B1 (KLKB1), angiotensin converting enzyme 2 (ACE2), and C1 esterase inhibitor (C1INH)] were analyzed. We detected dysregulated KKS in patients with COVID-19, characterized by an accumulation of BK1-8 in combination with decreased levels of BK. Accumulated BK1-8 was related to severity of patients with COVID-19. A multivariate logistic regression model retained BK1-8, BK, and D-dimer as independent predictor factors to intensive care unit (ICU) admission. A Youden’s optimal cutoff value of −0.352 was found for the multivariate model score with an accuracy of 92.9%. Multivariate model score-high group presented an odds ratio for ICU admission of 260.0. BK1-8 was related to inflammation, coagulation, and lymphopenia. Our data suggest that BK1-8/BK plasma concentration in combination with D-dimer levels might be retained as independent predictors for ICU admission in patients with COVID-19. Moreover, we reported KKS dysregulation in patients with COVID-19, which was related to disease severity by means of inflammation, hypercoagulation, and lymphopenia.
Collapse
Affiliation(s)
- Enrique Alfaro
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Elena Díaz-García
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Sara García-Tovar
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Ester Zamarrón
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Alberto Mangas
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Raúl Galera
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | | | - Rebeca Pérez-de-Diego
- Laboratory of Immunogenetics of Human Diseases, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain
| | | | - Francisco García-Río
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
- *Correspondence: Francisco García-Río, ; Carolina Cubillos-Zapata,
| | - Carolina Cubillos-Zapata
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- *Correspondence: Francisco García-Río, ; Carolina Cubillos-Zapata,
| |
Collapse
|
11
|
Ambrosino P, Bachetti T, D’Anna SE, Galloway B, Bianco A, D’Agnano V, Papa A, Motta A, Perrotta F, Maniscalco M. Mechanisms and Clinical Implications of Endothelial Dysfunction in Arterial Hypertension. J Cardiovasc Dev Dis 2022; 9:136. [PMID: 35621847 PMCID: PMC9146906 DOI: 10.3390/jcdd9050136] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 02/06/2023] Open
Abstract
The endothelium is composed of a monolayer of endothelial cells, lining the interior surface of blood and lymphatic vessels. Endothelial cells display important homeostatic functions, since they are able to respond to humoral and hemodynamic stimuli. Thus, endothelial dysfunction has been proposed as a key and early pathogenic mechanism in many clinical conditions. Given the relevant repercussions on cardiovascular risk, the complex interplay between endothelial dysfunction and systemic arterial hypertension has been a matter of study in recent years. Numerous articles have been published on this issue, all of which contribute to providing an interesting insight into the molecular mechanisms of endothelial dysfunction in arterial hypertension and its role as a biomarker of inflammation, oxidative stress, and vascular disease. The prognostic and therapeutic implications of endothelial dysfunction have also been analyzed in this clinical setting, with interesting new findings and potential applications in clinical practice and future research. The aim of this review is to summarize the pathophysiology of the relationship between endothelial dysfunction and systemic arterial hypertension, with a focus on the personalized pharmacological and rehabilitation strategies targeting endothelial dysfunction while treating hypertension and cardiovascular comorbidities.
Collapse
Affiliation(s)
- Pasquale Ambrosino
- Istituti Clinici Scientifici Maugeri IRCCS, Cardiac Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy;
| | - Tiziana Bachetti
- Istituti Clinici Scientifici Maugeri IRCCS, Scientific Direction, 27100 Pavia, Italy;
| | - Silvestro Ennio D’Anna
- Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy;
| | - Brurya Galloway
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (B.G.); (A.B.); (V.D.); (F.P.)
| | - Andrea Bianco
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (B.G.); (A.B.); (V.D.); (F.P.)
| | - Vito D’Agnano
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (B.G.); (A.B.); (V.D.); (F.P.)
| | - Antimo Papa
- Istituti Clinici Scientifici Maugeri IRCCS, Cardiac Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy;
| | - Andrea Motta
- Institute of Biomolecular Chemistry, National Research Council, 80078 Pozzuoli, Italy;
| | - Fabio Perrotta
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (B.G.); (A.B.); (V.D.); (F.P.)
| | - Mauro Maniscalco
- Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy;
- Department of Clinical Medicine and Surgery, “Federico II” University, 80131 Naples, Italy
| |
Collapse
|
12
|
Zeman M, Skałba W, Szymański P, Hadasik G, Żaworonkow D, Walczak DA, Czarniecka A. Risk factors for long-term survival in patients with ypN+ M0 rectal cancer after radical anterior resection. BMC Gastroenterol 2022; 22:141. [PMID: 35346064 PMCID: PMC8961971 DOI: 10.1186/s12876-022-02226-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 03/21/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Regional lymph node metastases are the main adverse prognostic factor in patients with rectal cancer without distant metastases. There are discrepancies, however, regarding additional risk factors in the group of ypN + M0 patients. The purpose of the study was to assess clinical and pathological factors affecting long-term oncological outcomes in the group of ypN + M0 patients after radical rectal anterior resection.
Methods
112 patients with ypN + M0 rectal cancer after neoadjuvant therapy and radical anterior resection were subject to a retrospective analysis. The effect of potential factors on survival was assessed with the use of Kaplan–Meier curves together with a log-rank test and multiple factor Cox proportional hazards model.
Results
In the multiple factor Cox analysis, adverse factors affecting disease-free survival (DFS) were: the use of angiotensin-converting enzyme inhibitors (ACEIs) (hazard ratio HR: 3.11, 95% CI 1.01–9.56, p = 0.047), presence of perineural invasion (HR: 7.27, 95% CI 2.74–19.3, p < 0.001) and occurrence of postoperative complications (HR: 6.79, 95% CI 2.09–22.11, p = 0.001), while a positive factor was the negative lymph node (NLN) count > 7 (HR: 0.33, 95% CI 0.12–0.88, p = 0.026). In the disease-specific survival (DSS) analysis, an adverse factor was the use of ACEIs (HR: 4.275, 95% CI 1.44–12.694, p = 0.009), while a positive effect was caused by NLN > 5 (HR: 0.22, 95% CI 0.082–0.586, p = 0.002).
Conclusions
The use of ACEIs may have a negative effect on long-term treatment outcomes in patients with ypN + M0 rectal cancer. In this group of patients, the NLN count seems to be an important prognostic factor, as well.
Collapse
|
13
|
Hillmeister P, Nagorka S, Gatzke N, Dülsner A, Li K, Dai M, Bondke Persson A, Lauxmann MA, Jaurigue J, Ritter O, Bramlage P, Buschmann E, Buschmann I. Angiotensin-converting enzyme inhibitors stimulate cerebral arteriogenesis. Acta Physiol (Oxf) 2022; 234:e13732. [PMID: 34555240 DOI: 10.1111/apha.13732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 12/20/2022]
Abstract
AIM Arteriogenesis constitutes the most efficient endogenous rescue mechanism in cases of cerebral ischaemia. The aim of this work was to investigate whether angiotensin-converting enzyme inhibitors (ACEi) stimulates, and angiotensin II receptor type 1 blockers (ARB) inhibits cerebral collateral growth by applying a three-vessel occlusion (3-VO) model in rat. METHODS Cerebral collateral growth was measured post 3-VO (1) by assessing blood flow using the cerebrovascular reserve capacity (CVRC) technique, and (2) by assessing vessel diameters in the posterior cerebral artery (PCA) via the evaluation of latex angiographies. A stimulatory effect on arteriogenesis was investigated for ACEi administration ± bradykinin receptor 1 (B1R) and 2 (B2R) blockers, and an inhibitory effect was analysed for ARB administration. Results were validated by immunohistochemical analysis and mechanistic data were collected by human umbilical vein endothelial cell (HUVEC) viability or scratch assay and monocyte (THP-1) migration assay. RESULTS An inhibitory effect of ARB on arteriogenesis could not be demonstrated. However, collateral growth measurements demonstrated a significantly increased CVRC and PCA diameters in the ACEi group. ACEi stimulates cell viability and migration, which could be partially reduced by additional administration of bradykinin receptor 1 inhibitor (B1Ri). ACEi inhibits the degradation of pro-arteriogenic bradykinin derivatives, but combined ACEi + B1Ri + B1Ri (BRB) treatment did not reverse the stimulatory effect. Yet, co-administration of ACEi + BRB enhances arteriogenesis and cell migration. CONCLUSION We demonstrate a potent stimulatory effect of ACEi on cerebral arteriogenesis in rats, presumable via B1R. However, results imply a pleiotropic and compensatory effect of ACEi on bradykinin receptor-stimulated arteriogenesis.
Collapse
Affiliation(s)
- Philipp Hillmeister
- Brandenburg Medical School Theodor Fontane (MHB) Deutsche Angiologie Zentrum Brandenburg‐Berlin (DAZB) Department for Angiology Center for Internal Medicine I Campus University Clinic Brandenburg Brandenburg an der Havel Germany
- Faculty of Health Sciences (FGW) Joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg the Brandenburg Medical School Theodor Fontane (MHB) University of Potsdam Brandenburg an der Havel Germany
| | | | - Nora Gatzke
- Brandenburg Medical School Theodor Fontane (MHB) Deutsche Angiologie Zentrum Brandenburg‐Berlin (DAZB) Department for Angiology Center for Internal Medicine I Campus University Clinic Brandenburg Brandenburg an der Havel Germany
| | | | - Kangbo Li
- Brandenburg Medical School Theodor Fontane (MHB) Deutsche Angiologie Zentrum Brandenburg‐Berlin (DAZB) Department for Angiology Center for Internal Medicine I Campus University Clinic Brandenburg Brandenburg an der Havel Germany
- Charité Universitätsmedizin Berlin Berlin Germany
| | - Mengjun Dai
- Brandenburg Medical School Theodor Fontane (MHB) Deutsche Angiologie Zentrum Brandenburg‐Berlin (DAZB) Department for Angiology Center for Internal Medicine I Campus University Clinic Brandenburg Brandenburg an der Havel Germany
- Charité Universitätsmedizin Berlin Berlin Germany
| | | | - Martin A. Lauxmann
- Brandenburg Medical School Theodor Fontane (MHB) Deutsche Angiologie Zentrum Brandenburg‐Berlin (DAZB) Department for Angiology Center for Internal Medicine I Campus University Clinic Brandenburg Brandenburg an der Havel Germany
- Brandenburg Medical School Theodor Fontane (MHB) Brandenburg Medical School (MHB) Theodor Fontane Institute for Biochemistry & Clinic for Nephrology Brandenburg an der Havel Germany
| | - Jonnel Jaurigue
- Brandenburg Medical School Theodor Fontane (MHB) Deutsche Angiologie Zentrum Brandenburg‐Berlin (DAZB) Department for Angiology Center for Internal Medicine I Campus University Clinic Brandenburg Brandenburg an der Havel Germany
| | - Oliver Ritter
- Brandenburg Medical School Theodor Fontane (MHB) Brandenburg Medical School (MHB) Theodor Fontane Institute for Biochemistry & Clinic for Nephrology Brandenburg an der Havel Germany
- Brandenburg Medical School Theodor Fontane (MHB) Department for Cardiology Center for Internal Medicine I Campus University Clinic Brandenburg Brandenburg an der Havel Germany
| | - Peter Bramlage
- Institute for Pharmacology and Preventive Medicine Cloppenburg Germany
| | - Eva Buschmann
- Department of Cardiology University Clinic Graz Graz Austria
| | - Ivo Buschmann
- Brandenburg Medical School Theodor Fontane (MHB) Deutsche Angiologie Zentrum Brandenburg‐Berlin (DAZB) Department for Angiology Center for Internal Medicine I Campus University Clinic Brandenburg Brandenburg an der Havel Germany
- Faculty of Health Sciences (FGW) Joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg the Brandenburg Medical School Theodor Fontane (MHB) University of Potsdam Brandenburg an der Havel Germany
| |
Collapse
|
14
|
Jiang F, Wang Y, Liu C, Zhang B, Wang E, Liu J, Zhang T. Egg White-Derived Peptides QVPLW and LCAY Inhibit the Activity of Angiotensin I-Converting Enzyme in Human Umbilical Vein Endothelial Cells by Suppressing Its Recruitment into Lipid Rafts. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:10350-10357. [PMID: 34448567 DOI: 10.1021/acs.jafc.1c04512] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
As a membrane protein, the activity of angiotensin I-converting enzyme (ACE) can be modulated via regulation of its localization in the cell membrane with food-derived peptides. This study aimed to explore the effect of egg white peptides on the cell membrane localization and activity of ACE in human umbilical vein endothelial cells. ACE activity was found to be related to lipid rafts by using methyl-β-cyclodextrin (MβCD). QVPLW and LCAY can inhibit ACE activity by preventing ACE recruitment into lipid rafts, with in situ IC50 values of 238.46 ± 11.35 μM and 31.55 ± 2.64 μM in the control groups, as well as 45.43 ± 6.15 μM and 34.63 ± 1.59 μM in the MβCD groups, respectively. QVPLW and LCAY may alter the cell membrane properties, including the fluidity, potential, and permeability, and eventually promote the transposition of ACE.
Collapse
Affiliation(s)
- Feng Jiang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Ying Wang
- College of Food Engineering, Jilin Engineering Normal University, Changchun 130052, People's Republic of China
| | - Chang Liu
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Biying Zhang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Erlei Wang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Jingbo Liu
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| | - Ting Zhang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, People's Republic of China
| |
Collapse
|
15
|
Li K, Zemmrich C, Bramlage P, Persson AB, Sacirovic M, Ritter O, Buschmann E, Buschmann I, Hillmeister P. Effect of ACEI and ARB treatment on nitric oxide-dependent endothelial function. VASA 2021; 50:413-422. [PMID: 34428929 DOI: 10.1024/0301-1526/a000971] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background: Angiotensin-converting-enzyme inhibitors (ACEI) and angiotensin II receptor blockers (ARB) are widely used as a first-line therapy for the treatment of cardiovascular disease. Here, ACEI modulate the bradykinin receptor (BDKRB1 and BDKRB2) system and NO-dependent endothelial function, thus determining cardiovascular health and regenerative arteriogenesis. The current study aims at evaluating nitric oxide-dependent endothelial function, and gene expression of bradykinin receptors in peripheral blood mononuclear cells (PBMC) from patients with ACEI or ARB treatment. Patients and methods: The WalkByLab has been established to screen cardiovascular patients for peripheral artery disease and coronary artery disease. In total 177 patients from WalkByLab with heterogenous disease and risk status were randomly selected, divided according to their medication history into the following groups: 1. ACEI group, 2. ARB group or 3. non-ACE/ARB group. Total plasma nitrite/nitrate (NO) levels were measured, endothelial function was evaluated by assessing flow meditated dilation (FMD). PBMC were isolated from peripheral whole blood, and gene expression (qRT-PCR) of bradykinin receptors and angiotensin converting enzyme were assessed. Results: Plasma total NO concentration in the ACEI group (24.66±16.28, µmol/l) was increased as compared to the ARB group (18.57±11.58, µmol/l, P=0.0046) and non-ACE/ARB group (16.83±8.64, µmol/l, P=0.0127) in patients between 40 to 90 years of age. However, FMD values (%) in the ACEI group (7.07±2.40, %) were similar as compared to the ARB (6.35±2.13, %) and non-ACE/ARB group (6.51±2.15, %), but significantly negatively correlated with age. Interestingly, BDKRB1 mRNA level was significantly higher and BDKRB2 mRNA level lower in the ACEI group (BDKRB1 3.88-fold±1.05, BDKRB2 0.22-fold±0.04) as compared to the non-ACE/ARB group (BDKRB1 1.00-fold±0.39, P<0.0001, BDKRB2 1.00-fold±0.45, P=0.0136). Conclusions: ACEI treatment enhances total nitrite/nitrate concentration, furthermore, upregulates BDKRB1 in PBMC, but downregulates BDKRB2 mRNA expression. FMD is a strong determinant of vascular aging and is sensitive to underlying heterogenous cardiovascular diseases.
Collapse
Affiliation(s)
- Kangbo Li
- Department for Angiology, Center for Internal Medicine I, Brandenburg Medical School Theodor Fontane, Campus University Clinic Brandenburg, Deutsches Angiologie Zentrum Brandenburg-Berlin (DAZB), Brandenburg an der Havel, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| | - Claudia Zemmrich
- Institute for Pharmacology and Preventive Medicine, Cloppenburg, Germany
| | - Peter Bramlage
- Institute for Pharmacology and Preventive Medicine, Cloppenburg, Germany
| | - Anja Bondke Persson
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| | - Mesud Sacirovic
- Department for Angiology, Center for Internal Medicine I, Brandenburg Medical School Theodor Fontane, Campus University Clinic Brandenburg, Deutsches Angiologie Zentrum Brandenburg-Berlin (DAZB), Brandenburg an der Havel, Germany
| | - Oliver Ritter
- Department for Cardiology, Center for Internal Medicine I, Brandenburg Medical School Theodor Fontane, Campus University Clinic Brandenburg, Brandenburg an der Havel, Germany.,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 Medical School Theodor Fontane, Germany
| | - Eva Buschmann
- Department of Cardiology, University Clinic Graz, Austria
| | - Ivo Buschmann
- Department for Angiology, Center for Internal Medicine I, Brandenburg Medical School Theodor Fontane, Campus University Clinic Brandenburg, Deutsches Angiologie Zentrum Brandenburg-Berlin (DAZB), Brandenburg an der Havel, Germany.,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 Medical School Theodor Fontane, Germany
| | - Philipp Hillmeister
- Department for Angiology, Center for Internal Medicine I, Brandenburg Medical School Theodor Fontane, Campus University Clinic Brandenburg, Deutsches Angiologie Zentrum Brandenburg-Berlin (DAZB), Brandenburg an der Havel, Germany.,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 Medical School Theodor Fontane, Germany
| |
Collapse
|
16
|
Aliska G, Dhuha A, Rahmatini R, Hamdani R, Afriani N, Tofrizal T, Ali H, Pratiwi N, Nuranisyah V, Mahata LE, Kurnia D. Ramipril Increases Adma Concentration in Acute Myocardial Infarction in Rats Induced by Isoproterenol. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
In experimental animals, the induction of isoproterenol which is a synthetic of catecholamine, can cause acute myocardial infarction where the pathophysiology and morphology are the same as myocardial infarction in humans. Isoproterenol induction will increase oxidative stress, which will damage the enzyme dimethylarginine dimethylaminohydrolase (DDAH), thus causing asymmetric dimethylarginine (ADMA) levels to increase in circulation. Increased levels of ADMA will inhibit the activity of the enzyme nitric oxide synthase, which results in decreased nitric oxide resulting in endothelial damage. This study aims to determine the effect of Ramipril on asymmetric dimethylarginine (ADMA) levels in rats (Rattus norvegicus) Wistar strain with acute myocardial infarction.
Eighteen male Wistar rats (150-250 g) were randomly allocated into three groups: negative control group, positive control, and treatment group. The treatment group was pretreated with Ramipril at dose 3 mg/kg BW orally for seven days. Acute myocardial infarction was induced in positive control groups and treatment groups by subcutaneous injection of isoproterenol (85 mg/kg BW) for two consecutive days. Twenty-four hours after the last administration, rats from all groups were anesthetized and sacrificed for blood sample collection to evaluate the level of Asymmetric Dimethylarginine with Enzym-linked Immunosorbent Assay (ELISA) method. The result showed that ADMA levels were increased in the treatment group after pretreated with Ramipril. This study concluded that pretreatment with Ramipril increased ADMA concentration in acute myocardial infarction rats induced by isoproterenol.
Collapse
|
17
|
de Souza Maciel I, Azevedo VM, Oliboni P, Campos MM. Blockade of the kinin B 1 receptor counteracts the depressive-like behaviour and mechanical allodynia in ovariectomised mice. Behav Brain Res 2021; 412:113439. [PMID: 34197868 DOI: 10.1016/j.bbr.2021.113439] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 05/31/2021] [Accepted: 06/25/2021] [Indexed: 12/27/2022]
Abstract
Menopause is related to a decline in ovarian oestrogen production, affecting the perception of the somatosensory stimuli, changing the immune-inflammatory systems, and triggering depressive symptoms. It has been demonstrated that the inhibition of the kinin B1 and B2 receptors (B1R and B2R) prevented the depressive-like behaviour and the mechanical allodynia that was induced by immune-inflammatory mediators in mice. However, there is no evidence regarding the role of the kinin receptors in the depressive-like and nociceptive behaviour in female mice that were subjected to bilateral ovariectomy (OVX). This study has shown that the OVX mice developed time-related mechanical allodynia, together with an increased immobility time as indicative of depression. Both of these changes were reduced by the genetic deletion of B1R, or by the pharmacological blockade of the selective kinin B1R antagonist R-715 (acute, i.p.). The genetic deletion or the pharmacological inhibition of B2R (HOE 140, i.p.) did not prevent the OVX-elicited behavioural changes. The data has suggested a particular modulation of kinin B1R in the nociceptive and depressive-like behaviour in the OVX mice. The selective inhibition of the B1R receptor may be a new pharmacological target for treating pain and depression symptoms in women during the perimenopause/menopause period.
Collapse
Affiliation(s)
- Izaque de Souza Maciel
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Escola de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil.
| | - Vanessa Machado Azevedo
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Patricia Oliboni
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maria Martha Campos
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
18
|
Sánchez-López F, Robles-Olvera VJ, Hidalgo-Morales M, Tsopmo A. Angiotensin-I converting enzyme inhibitory activity of Amaranthus hypochondriacus seed protein hydrolysates produced with lactic bacteria and their peptidomic profiles. Food Chem 2021; 363:130320. [PMID: 34146770 DOI: 10.1016/j.foodchem.2021.130320] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/31/2021] [Accepted: 06/04/2021] [Indexed: 11/26/2022]
Abstract
The aim of this work was to determine the in vitro antihypertensive activities of lactobacillus (L. plantarum and L. helveticus) prepared amaranth protein hydrolysates, to determine the contribution of zinc, and to identify peptides. Depending on the bacteria species and the duration of the hydrolysis, up to 45.9% inhibition of angiotensin converting enzyme (ACE) was obtained. Size separation of the most active hydrolysates to yield < 1, <3-1, <3, <10-3 and < 10 kDa fractions enhanced ACE inhibition by 2-fold. A mixed mechanism of inhibition is proposed due to low correlation of ACE and zinc chelation. Thirty-six peptides were identified in the fractions using tandem mass spectrometry. A bioinformatic analysis showed the presence of encrypted fragments such as GVSEE or VNVDDPSK with known ACE-inhibitory properties. In conclusion, lactic acid bacteria proteases released peptides from amaranth proteins with ACE-inhibitory properties that were related to the presence of peptides with known or predicted ACE-inhibitor motifs.
Collapse
Affiliation(s)
- Fabiola Sánchez-López
- Tecnológico Nacional de México-Instituto Tecnológico de Veracruz-UNIDA, M.A. de Quevedo #2779, Col. Formando Hogar, Veracruz 91897, Mexico
| | - Víctor J Robles-Olvera
- Tecnológico Nacional de México-Instituto Tecnológico de Veracruz-UNIDA, M.A. de Quevedo #2779, Col. Formando Hogar, Veracruz 91897, Mexico
| | - Madeleine Hidalgo-Morales
- Tecnológico Nacional de México-Instituto Tecnológico de Veracruz-UNIDA, M.A. de Quevedo #2779, Col. Formando Hogar, Veracruz 91897, Mexico
| | - Apollinaire Tsopmo
- Food Science Program, Department of Chemistry, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
19
|
Ruggenenti P, Podestà MA, Trillini M, Perna A, Peracchi T, Rubis N, Villa D, Martinetti D, Cortinovis M, Ondei P, Condemi CG, Guastoni CM, Meterangelis A, Granata A, Mambelli E, Pasquali S, Genovesi S, Pieruzzi F, Bertoli SV, Del Rosso G, Garozzo M, Rigotti A, Pozzi C, David S, Daidone G, Mingardi G, Mosconi G, Galfré A, Romei Longhena G, Pacitti A, Pani A, Hidalgo Godoy J, Anders HJ, Remuzzi G. Ramipril and Cardiovascular Outcomes in Patients on Maintenance Hemodialysis: The ARCADIA Multicenter Randomized Controlled Trial. Clin J Am Soc Nephrol 2021; 16:575-587. [PMID: 33782036 PMCID: PMC8092055 DOI: 10.2215/cjn.12940820] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/15/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND OBJECTIVES Renin-angiotensin system (RAS) inhibitors reduce cardiovascular morbidity and mortality in patients with CKD. We evaluated the cardioprotective effects of the angiotensin-converting enzyme inhibitor ramipril in patients on maintenance hemodialysis. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS In this phase 3, prospective, randomized, open-label, blinded end point, parallel, multicenter trial, we recruited patients on maintenance hemodialysis with hypertension and/or left ventricular hypertrophy from 28 Italian centers. Between July 2009 and February 2014, 140 participants were randomized to ramipril (1.25-10 mg/d) and 129 participants were allocated to non-RAS inhibition therapy, both titrated up to the maximally tolerated dose to achieve predefined target BP values. The primary efficacy end point was a composite of cardiovascular death, myocardial infarction, or stroke. Secondary end points included the single components of the primary end point, new-onset or recurrence of atrial fibrillation, hospitalizations for symptomatic fluid overload, thrombosis or stenosis of the arteriovenous fistula, and changes in cardiac mass index. All outcomes were evaluated up to 42 months after randomization. RESULTS At comparable BP control, 23 participants on ramipril (16%) and 24 on non-RAS inhibitor therapy (19%) reached the primary composite end point (hazard ratio, 0.93; 95% confidence interval, 0.52 to 1.64; P=0.80). Ramipril reduced cardiac mass index at 1 year of follow-up (between-group difference in change from baseline: -16.3 g/m2; 95% confidence interval, -29.4 to -3.1), but did not significantly affect the other secondary outcomes. Hypotensive episodes were more frequent in participants allocated to ramipril than controls (41% versus 12%). Twenty participants on ramipril and nine controls developed cancer, including six gastrointestinal malignancies on ramipril (four were fatal), compared with none in controls. CONCLUSIONS Ramipril did not reduce the risk of major cardiovascular events in patients on maintenance hemodialysis. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER ARCADIA, NCT00985322 and European Union Drug Regulating Authorities Clinical Trials Database number 2008-003529-17.
Collapse
Affiliation(s)
- Piero Ruggenenti
- Department of Renal Medicine, Clinical Research Centre for Rare Diseases “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Manuel Alfredo Podestà
- Department of Renal Medicine, Clinical Research Centre for Rare Diseases “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Matias Trillini
- Department of Renal Medicine, Clinical Research Centre for Rare Diseases “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Annalisa Perna
- Department of Renal Medicine, Clinical Research Centre for Rare Diseases “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Tobia Peracchi
- Department of Renal Medicine, Clinical Research Centre for Rare Diseases “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Nadia Rubis
- Department of Renal Medicine, Clinical Research Centre for Rare Diseases “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Davide Villa
- Department of Renal Medicine, Clinical Research Centre for Rare Diseases “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Davide Martinetti
- Department of Renal Medicine, Clinical Research Centre for Rare Diseases “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Monica Cortinovis
- Department of Renal Medicine, Clinical Research Centre for Rare Diseases “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Patrizia Ondei
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Carmela Giuseppina Condemi
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Carlo Maria Guastoni
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Ovest Milanese, Ospedali di Legnano e Magenta, Milano, Italy
| | - Agnese Meterangelis
- Unit of Nephrology and Dialysis, Policlinico San Pietro, Ponte San Pietro, Bergamo, Italy
| | - Antonio Granata
- Unit of Nephrology and Dialysis, Azienda Ospedaliera per l'Emergenza “Cannizzaro,” Catania, Italy
| | - Emanuele Mambelli
- Unit of Nephrology, Dialysis and Hypertension, Azienda Ospedaliero-Universitaria di Bologna Policlinico S. Orsola-Malpighi, Bologna, Italy
| | - Sonia Pasquali
- Unit of Nephrology and Dialysis, Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | - Simonetta Genovesi
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Monza, Ospedale San Gerardo, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy
| | - Federico Pieruzzi
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Monza, Ospedale San Gerardo, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy
| | - Silvio Volmer Bertoli
- Unit of Nephrology and Dialysis, Istituto di Ricovero e Cura a Carattere Scientifico MultiMedica, Sesto San Giovanni, Milano, Italy
| | - Goffredo Del Rosso
- Unit of Nephrology and Dialysis, Ospedale Giuseppe Mazzini, Teramo, Italy
| | - Maurizio Garozzo
- Unit of Nephrology and Dialysis, Presidio Ospedaliero S. Marta e S. Venera, Acireale, Catania, Italy
| | - Angelo Rigotti
- Unit of Nephrology and Dialysis, Ospedale Infermi, Rimini, Italy
| | - Claudio Pozzi
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Nord Milano-Ospedale Bassini, Cinisello Balsamo, Milano, Italy
| | - Salvatore David
- Department of Nephrology and Dialysis, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Giuseppe Daidone
- Unit of Nephrology and Dialysis, Ospedale Umberto I, Siracusa, Italy
| | - Giulio Mingardi
- Unit of Nephrology and Dialysis, Humanitas Gavazzeni, Bergamo, Italy
| | - Giovanni Mosconi
- Unit of Nephrology and Dialysis, Ospedale “Morgagni-Pierantoni,” Forlì, Italy
| | - Andrea Galfré
- Unit of Nephrology and Dialysis, Azienda Sanitaria Locale 8, Cagliari, Italy
| | - Giorgio Romei Longhena
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Rhodense-Ospedale Garbagnate Milanese, Milano, Italy
| | - Alfonso Pacitti
- Unit of Nephrology and Dialysis, Azienda Ospedaliera Santa Croce e Carle, Cuneo, Italy
| | - Antonello Pani
- Unit of Nephrology and Dialysis, Department of Reproduction, Genitourinary and Kidney Disease and Kidney Transplantation, Azienda Ospedaliera Brotzu, Cagliari, Italy
| | - Jorge Hidalgo Godoy
- Department of Renal Medicine, Clinical Research Centre for Rare Diseases “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
- Institute of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig Maximilians Universität Munich, Munich, Germany
| | - Giuseppe Remuzzi
- Department of Renal Medicine, Clinical Research Centre for Rare Diseases “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | | |
Collapse
|
20
|
Brusco I, Justino AB, Silva CR, Scussel R, Machado-de-Ávila RA, Oliveira SM. Inhibitors of angiotensin I converting enzyme potentiate fibromyalgia-like pain symptoms via kinin receptors in mice. Eur J Pharmacol 2021; 895:173870. [PMID: 33476653 DOI: 10.1016/j.ejphar.2021.173870] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 12/18/2020] [Accepted: 01/11/2021] [Indexed: 01/06/2023]
Abstract
Fibromyalgia is a potentially disabling chronic disease, characterized by widespread pain and a range of comorbidities such as hypertension. Among the mechanisms involved in fibromyalgia-like pain symptoms are kinins and their B1 and B2 receptors. Moreover, angiotensin I converting enzyme (ACE) inhibitors, commonly used as antihypertensive drugs, can enhance pain by blocking the degradation of peptides such as substance P and bradykinin, besides enhancing kinin receptors signalling. We investigated the effect of ACE inhibitors on reserpine-induced fibromyalgia-like pain symptoms and the involvement of kinins in this effect in mice. Nociceptive parameters (mechanical and cold allodynia and overt nociception) were evaluated after ACE inhibitors administration in mice previously treated with reserpine. The role of kinin B1 and B2 receptors was investigated using pharmacological antagonism. Additionally, bradykinin levels, as well as the activity of ACE and kininase I, were measured in the sciatic nerve, spinal cord and cerebral cortex of the mice. The ACE inhibitors enalapril and captopril enhanced reserpine-induced mechanical allodynia, and this increase was prevented by kinin B1 and B2 receptor antagonists. Substance P and bradykinin caused overt nociception and increased mechanical allodynia in animals treated with reserpine. Reserpine plus ACE inhibitors increased bradykinin-related peptide levels and inhibited ACE activity in pain modulation structures. Since hypertension is a frequent comorbidity affecting fibromyalgia patients, hypertension treatment with ACE inhibitors in these patients should be reviewed once this could enhance fibromyalgia-like pain symptoms. Thus, the treatment of hypertensive patients with fibromyalgia could include other classes of antihypertensive drugs, different from ACE inhibitors.
Collapse
Affiliation(s)
- Indiara Brusco
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Allisson Benatti Justino
- Graduate Program in Genetics and Biochemistry, Biotechnology Institute, Federal University of Uberlandia, Uberlandia, MG, Brazil
| | - Cássia Regina Silva
- Graduate Program in Genetics and Biochemistry, Biotechnology Institute, Federal University of Uberlandia, Uberlandia, MG, Brazil
| | - Rahisa Scussel
- Laboratory of Cellular and Molecular Biology, Health Sciences Academic Unit, University of Extreme South Catarinense, Criciuma, SC, Brazil
| | | | - Sara Marchesan Oliveira
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
21
|
Nunes MA, Toricelli M, Schöwe NM, Malerba HN, Dong-Creste KE, Farah DMAT, De Angelis K, Irigoyen MC, Gobeil F, Araujo Viel T, Buck HS. Kinin B2 Receptor Activation Prevents the Evolution of Alzheimer's Disease Pathological Characteristics in a Transgenic Mouse Model. Pharmaceuticals (Basel) 2020; 13:ph13100288. [PMID: 33019732 PMCID: PMC7601323 DOI: 10.3390/ph13100288] [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: 08/12/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 12/02/2022] Open
Abstract
Background: Alzheimer’s disease is mainly characterized by remarkable neurodegeneration in brain areas related to memory formation. This progressive neurodegeneration causes cognitive impairment, changes in behavior, functional disability, and even death. Our group has demonstrated changes in the kallikrein–kinin system (KKS) in Alzheimer’s disease (AD) experimental models, but there is a lack of evidence about the role of the KKS in Alzheimer’s disease. Aim: In order to answer this question, we evaluated the potential of the kinin B2 receptors (BKB2R) to modify AD characteristics, particularly memory impairment, neurodegeneration, and Aβ peptide deposition. Methods: To assess the effects of B2, we used transgenic Alzheimer’s disease mice treated with B2 receptor (B2R) agonists and antagonists, and performed behavioral and biochemical tests. In addition, we performed organotypic hippocampal culture of wild-type (WT) and transgenic (TG) animals, where the density of cytokines, neurotrophin BDNF, activated astrocyte marker S100B, and cell death were analyzed after treatments. Results: Treatment with the B2R agonist preserved the spatial memory of transgenic mice and decreased amyloid plaque deposition. In organotypic hippocampal culture, treatment with B2R agonist decreased cell death, neuroinflammation, and S100B levels, and increased BDNF release. Conclusions: Our results indicate that the kallikrein–kinin system plays a beneficial role in Alzheimer’s disease through B2R activation. The use of B2R agonists could, therefore, be a possible therapeutic option for patients diagnosed with Alzheimer’s disease.
Collapse
Affiliation(s)
- Marielza Andrade Nunes
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo 01221-020, Brazil; (M.A.N.); (M.T.); (K.E.D.-C.)
| | - Mariana Toricelli
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo 01221-020, Brazil; (M.A.N.); (M.T.); (K.E.D.-C.)
| | - Natalia Mendes Schöwe
- School of Arts, Sciences and Humanities, University of Sao Paulo, Sao Paulo 03828-080, Brazil; (N.M.S.); (H.N.M.); (T.A.V.)
| | - Helena Nascimento Malerba
- School of Arts, Sciences and Humanities, University of Sao Paulo, Sao Paulo 03828-080, Brazil; (N.M.S.); (H.N.M.); (T.A.V.)
| | - Karis Ester Dong-Creste
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo 01221-020, Brazil; (M.A.N.); (M.T.); (K.E.D.-C.)
| | - Daniela Moura Azevedo Tuma Farah
- Heart Institute (Incor), Hypertension Unit, University of Sao Paulo, Sao Paulo 05403-900, Brazil; (D.M.A.T.F.); (M.C.I.)
- Department of Physiology, Federal University of São Paulo (UNIFESP), Sao Paulo 04023-901, Brazil;
| | - Katia De Angelis
- Department of Physiology, Federal University of São Paulo (UNIFESP), Sao Paulo 04023-901, Brazil;
- Translational Physiology Laboratory, Universidade Nove de Julho (UNINOVE), Sao Paulo 01504-001, Brazil
| | - Maria Claudia Irigoyen
- Heart Institute (Incor), Hypertension Unit, University of Sao Paulo, Sao Paulo 05403-900, Brazil; (D.M.A.T.F.); (M.C.I.)
| | - Fernand Gobeil
- Department of Pharmacology and Physiology, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Tânia Araujo Viel
- School of Arts, Sciences and Humanities, University of Sao Paulo, Sao Paulo 03828-080, Brazil; (N.M.S.); (H.N.M.); (T.A.V.)
| | - Hudson Sousa Buck
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo 01221-020, Brazil; (M.A.N.); (M.T.); (K.E.D.-C.)
- Correspondence: ; Tel./Fax: +55-11-3367-7790
| |
Collapse
|
22
|
Gouda AS, Mégarbane B. Snake venom-derived bradykinin-potentiating peptides: A promising therapy for COVID-19? Drug Dev Res 2020; 82:38-48. [PMID: 32761647 PMCID: PMC7436322 DOI: 10.1002/ddr.21732] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/23/2022]
Abstract
The severe acute respiratory syndrome coronavirus‐2 (SARS‐COV‐2), a novel coronavirus responsible for the recent infectious pandemic, is known to downregulate angiotensin‐converting enzyme‐2 (ACE2). Most current investigations focused on SARS‐COV‐2‐related effects on the renin–angiotensin system and especially the resultant increase in angiotensin II, neglecting its effects on the kinin–kallikrein system. SARS‐COV‐2‐induced ACE2 inhibition leads to the augmentation of bradykinin 1‐receptor effects, as ACE2 inactivates des‐Arg9‐bradykinin, a bradykinin metabolite. SARS‐COV‐2 also decreases bradykinin 2‐receptor effects as it affects bradykinin synthesis by inhibiting cathepsin L, a kininogenase present at the site of infection and involved in bradykinin production. The physiologies of both the renin–angiotensin and kinin–kallikrein system are functionally related suggesting that any intervention aiming to treat SARS‐COV‐2‐infected patients by triggering one system but ignoring the other may not be adequately effective. Interestingly, the snake‐derived bradykinin‐potentiating peptide (BPP‐10c) acts on both systems. BPP‐10c strongly decreases angiotensin II by inhibiting ACE, increasing bradykinin‐related effects on the bradykinin 2‐receptor and increasing nitric oxide‐mediated effects. Based on a narrative review of the literature, we suggest that BPP‐10c could be an optimally effective option to consider when aiming at developing an anti‐SARS‐COV‐2 drug.
Collapse
Affiliation(s)
- Ahmed S Gouda
- National Egyptian Center for Toxicological Researches, Faculty of Medicine, University of Cairo, Cairo, Egypt
| | - Bruno Mégarbane
- Department of Medical and Toxicological Critical Care, Lariboisière Hospital, University of Paris, INSERM UMRS-1144, Paris, France
| |
Collapse
|
23
|
De Oliveira TV, Guimarães AP, Bressan GC, Maia ER, Coimbra JSDR, Polêto MD, De Oliveira EB. Structural and molecular bases of angiotensin-converting enzyme inhibition by bovine casein-derived peptides: an in silico molecular dynamics approach. J Biomol Struct Dyn 2020; 39:1386-1403. [PMID: 32066337 DOI: 10.1080/07391102.2020.1730243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The angiotensin-converting enzyme (ACE) plays a key role in blood pressure regulation process, and its inhibition is one of the main drug targets for the treatment of hypertension. Though various peptides from milk proteins are well-known for their ACE-inhibitory capacity, research devoted to understand the molecular bases of such property remain scarce, specifically for such peptides. Therefore, in this work, computational molecular docking and molecular dynamics calculations were performed to enlighten the intermolecular interactions involved in ACE inhibition by six different casein-derived peptides (FFVAPFPEVFGK, FALPQYLK, ALNEINQFYQK, YLGYLEQLLR, HQGLPQEVLNENLLR and NAVPITPTLNR). Two top ranked docking poses for each peptide (one with N- and the other C-terminal peptide extremity oriented towards the ACE active site) were selected for dynamic simulations (50 ns; GROMOS53A6 force field), and the results were correlated to in vitro ACE inhibition capacity. Two molecular features appeared to be essential for peptides to present high ACE inhibition capacity in vitro: i) to interact with the S1 active site residues (Ala354, Glu384, and Tyr523) by hydrogen bonds; ii) to interact with Zn2+ coordinated residues (His383, His387, and Glu411) by short-lenght hydrogen bonds, as observed in the cases of ALNEINQFYQK (IACE = 80.7%), NAVPITPTLNR (IACE = 80.7%), and FALPQYLK (IACE = 79.0%). Regardless of the temporal stability of these strong interactions, they promoted some disruption of Zn2+ tetrahedral coordination during the molecular dynamics trajectories, and were pointed as the main reason for the greatest ACE inhibition by these peptides. On the other hand, peptides with intermediate inhibition capacity (50% < IACE < 45%) interacted mainly by weaker interactions (e.g.: electrostatic and hydrophobic) with the Zn2+ coordinated residues, and were not able to change significantly its tetrahedral coordination structure. These findings may: i) assist the discrimination in silico of "good" and "bad" ACE-inhibitory peptides from other food sources, and/or ii) aid in designing de novo new molecules with ACE-inhibitory capacity. Communicated by Ramaswamy Sarma.
Collapse
Affiliation(s)
| | - Ana Paula Guimarães
- Departamento de Química (DEQ), Universidade Federal de Viçosa (UFV), Viçosa, MG, Brazil
| | - Gustavo Costa Bressan
- Departamento de Bioquímica e Biologia Molecular (DBB), Universidade Federal de Viçosa (UFV), Viçosa, MG, Brazil
| | - Elaine Rose Maia
- Laboratório de Estudos Estruturais Moleculares (LEEM), Instituto de Química, Universidade de Brasília, Brasília, DF, Brazil
| | | | - Marcelo Depólo Polêto
- Departamento de Biologia Geral (DBG), Universidade Federal de Viçosa (UFV), Viçosa, MG, Brazil
| | | |
Collapse
|
24
|
Song TJ, Kim BS, Chu MK. Therapeutic role of melatonin in migraine prophylaxis: Is there a link between sleep and migraine? PROGRESS IN BRAIN RESEARCH 2020; 255:343-369. [DOI: 10.1016/bs.pbr.2020.05.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/12/2020] [Accepted: 05/01/2020] [Indexed: 12/13/2022]
|
25
|
Majid A, Priyadarshini C G P. Millet derived bioactive peptides: A review on their functional properties and health benefits. Crit Rev Food Sci Nutr 2019; 60:3342-3351. [DOI: 10.1080/10408398.2019.1686342] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Abdul Majid
- Department of Molecular Nutrition, CSIR - Central Food Technological Research Institute, Mysore, Karnataka, India
| | - Poornima Priyadarshini C G
- Department of Molecular Nutrition, CSIR - Central Food Technological Research Institute, Mysore, Karnataka, India
| |
Collapse
|
26
|
Kempe S, Fois G, Brunner C, Hoffmann TK, Hahn J, Greve J. Bradykinin signaling regulates solute permeability and cellular junction organization in lymphatic endothelial cells. Microcirculation 2019; 27:e12592. [PMID: 31550055 DOI: 10.1111/micc.12592] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 09/11/2019] [Accepted: 09/18/2019] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Determine the effect of bradykinin on solute permeability and cellular junctional proteins in human dermis microvascular endothelial cells. METHODS Cells were characterized by immunofluorescence and fluorescence-activated cell sorting. Macromolecular transport of dextran and albumin was monitored. Junctional protein expression and phosphorylation were determined by immunoblot analyses. Intracellular calcium and cAMP levels were evaluated. Target gene expression at mRNA and protein levels was determined. RESULTS Human dermis microvascular endothelial cells comprised 97% lymphatic endothelial cells. Bradykinin increased the permeability to dextran in a concentration-dependent manner, while reduced the permeability to albumin. Bradykinin treatment down-regulated VE-cadherin expression and affected its phosphorylation status at Tyr731. It also down-regulated claudin-5 expression at the transcriptional level through bradykinin-2-receptor signaling. An increase in the intracellular calcium levels and a reduction in the cAMP concentration were associated effects. Finally, bradykinin induced the up-regulation of vascular endothelial growth factor-C protein which was found increased in BK-induced human dermis microvascular endothelial cells culture supernates. CONCLUSIONS Human dermis microvascular endothelial cells represent a model of lymphatic endothelial cells, in which bradykinin-2-receptor is expressed. Bradykinin-induced bradykinin-2-receptor signaling through intracellular calcium mobilization and reduction in cAMP levels, triggered changes in solute permeability and cellular junction expression. It further up-regulated vascular endothelial growth factors-C protein expression, which is a key modulator of lymphatic vessels function and lymphangiogenesis.
Collapse
Affiliation(s)
- Sybille Kempe
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Giorgio Fois
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Cornelia Brunner
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Thomas K Hoffmann
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Janina Hahn
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Jens Greve
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
27
|
Brusco I, Justino AB, Silva CR, Fischer S, Cunha TM, Scussel R, Machado-de-Ávila RA, Ferreira J, Oliveira SM. Kinins and their B1 and B2 receptors are involved in fibromyalgia-like pain symptoms in mice. Biochem Pharmacol 2019; 168:119-132. [DOI: 10.1016/j.bcp.2019.06.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022]
|
28
|
Zhao W, Xue S, Yu Z, Ding L, Li J, Liu J. Novel ACE inhibitors derived from soybean proteins using in silico and in vitro studies. J Food Biochem 2019; 43:e12975. [PMID: 31489673 DOI: 10.1111/jfbc.12975] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 12/18/2022]
Abstract
The widespread application of soybean-derived peptides is currently limited due to the challenges in the identification of peptides. In the present work, in silico and in vitro analysis were applied to identify ACE inhibitory tri-peptides from soybean protein. The soybean protein was cleaved by PeptideCutter. Then, unknown tri-peptides were selected to solubility estimation and ADME prediction. Subsequently, Discovery Studio was applied to evaluate the interaction mechanism between ACE and tri-peptides. Finally, in vitro activity of theoretical ACE inhibitory tri-peptides was verified by RP-HPLC method. As a result, DMG was selected as a potent ACE inhibitory peptide. Cell experiment showed that DMG had no cytotoxic effects on HEK-293 cells. And molecular docking results indicated that DMG contacted well with ACE's active sites (Gln281, His353, Ala354, Glu384, Lys511, His513, and Tyr520). Furthermore, DMG could exert potent activity against ACE, with IC50 value of 3.95 ± 0.11 mM. PRACTICAL APPLICATIONS: Present research showed soybean is a potential protein resource to obtain ACE inhibitory peptides. Simultaneously, virtual screening method is a feasible way to substitute for classical method in emerging nutritional fields. What's more, present study provides a theoretical basis for industrial research on foodstuff for ACE inhibitory peptides without side effects.
Collapse
Affiliation(s)
- Wenzhu Zhao
- College of Food Science and Engineering, Bohai University, Jinzhou, P.R. China
| | - Siyu Xue
- College of Food Science and Engineering, Bohai University, Jinzhou, P.R. China
| | - Zhipeng Yu
- College of Food Science and Engineering, Bohai University, Jinzhou, P.R. China
| | - Long Ding
- Lab of Nutrition and Functional Food, Jilin University, Changchun, P.R. China
| | - Jianrong Li
- College of Food Science and Engineering, Bohai University, Jinzhou, P.R. China
| | - Jingbo Liu
- Lab of Nutrition and Functional Food, Jilin University, Changchun, P.R. China
| |
Collapse
|
29
|
Multifunctionality of lunasin and peptides released during its simulated gastrointestinal digestion. Food Res Int 2019; 125:108513. [PMID: 31554062 DOI: 10.1016/j.foodres.2019.108513] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/17/2019] [Accepted: 06/21/2019] [Indexed: 12/14/2022]
Abstract
Oxidative stress, inflammation, and hypertension are recognized risk factors for non-communicable diseases. Because of the preventable character of these factors, the searching of dietary compounds with counteracting effects against them would provide a new framework for the development of novel multifunctional foods or nutraceuticals. Lunasin is a naturally occurring soybean peptide with chemopreventive and anti-inflammatory properties. Upon oral intake, lunasin is susceptible to the action of digestive enzymes during its transit through gastrointestinal tract. In spite of its cleavage into smaller peptides, these fragments have been suggested to contribute on the health beneficial effects attributed to lunasin. To confirm this hypothesis, the multifunctionality of lunasin derived-fragments was investigated. In vitro, peptides corresponding to the N-terminal and central regions of lunasin were demonstrated to inhibit angiotensin converting enzyme and to scavenge peroxyl and 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulphonic acid (ABTS) radicals. Moreover, lunasin and fragments released during its gastrointestinal digestion exerted potent protective effects on cell viability and oxidative status in macrophages RAW264.7 challenged with chemicals tert-butylhydroperoxide and hydrogen peroxide. These peptides were also able to reduce the nitric oxide production in pro-inflammatory lipopolysaccharide-induced macrophages. These results confirm the promising role of lunasin and its derived-fragments as protective agents against oxidative damage and inflammation-associated diseases.
Collapse
|
30
|
Deng T, Xie X, Duan J, Chen M. Di-(2-ethylhexyl) phthalate induced an increase in blood pressure via activation of ACE and inhibition of the bradykinin-NO pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 247:927-934. [PMID: 30823347 DOI: 10.1016/j.envpol.2019.01.099] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/20/2019] [Accepted: 01/24/2019] [Indexed: 06/09/2023]
Abstract
Epidemiological studies and animal experiments have suggested that exposure to Di-(2-ethylhexyl) phthalate (DEHP) is strongly associated with an increase in blood pressure. However, the mechanisms that result in the detrimental effects of DEHP exposure on blood pressure are unclear. In our study, mice were orally exposed to DEHP dosages of 0.1, 1, 10 mg/kg/day for 6 weeks. The results showed that DEHP could induce a significant increase in systolic blood pressure (SBP) and heart rate, and a significant thickening of the ventricular wall. To explore the underlying mechanism, we measured the level of: angiotensin converting enzyme (ACE); bradykinin B2 receptor (BK2R); endothelial nitric oxide synthase (eNOS); bradykinin and Ca2+ in cardiac cytoplasm as well as in serum nitric oxide (NO). The results suggested that DEHP could induce an increase in ACE levels, and a decrease in bradykinin levels. Moreover, BK2R, Ca2+, eNOS and NO decreased when mice were exposed to 10 mg/kg/day DEHP. Interestingly, 5 mg/kg/day angiotensin converting enzyme inhibitor (ACEI) treatment inhibited the increase in blood pressure, and inhibited the decrease in the levels of BK2R, Ca2+, eNOS, and NO, that were induced by DEHP exposure. Our results suggest that DEHP might increase blood pressure by activating ACE expression, and inhibiting the bradykinin-NO pathway.
Collapse
Affiliation(s)
- Ting Deng
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei, 430079, China
| | - Xiaoman Xie
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei, 430079, China
| | - Jiufei Duan
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei, 430079, China
| | - Mingqing Chen
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei, 430079, China.
| |
Collapse
|
31
|
Marceau F, Bawolak MT, Fortin JP, Morissette G, Roy C, Bachelard H, Gera L, Charest-Morin X. Bifunctional ligands of the bradykinin B 2 and B 1 receptors: An exercise in peptide hormone plasticity. Peptides 2018; 105:37-50. [PMID: 29802875 DOI: 10.1016/j.peptides.2018.05.007] [Citation(s) in RCA: 14] [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: 03/22/2018] [Revised: 05/14/2018] [Accepted: 05/15/2018] [Indexed: 12/24/2022]
Abstract
Kinins are the small and fragile hydrophilic peptides related to bradykinin (BK) and derived from circulating kininogens via the action of kallikreins. Kinins bind to the preformed and widely distributed B2 receptor (B2R) and to the inducible B1 receptor (B1R). B2Rs and B1Rs are related G protein coupled receptors that possess natural agonist ligands of nanomolar affinity (BK and Lys BK for B2Rs, Lys-des-Arg9-BK for B1R). Decades of structure-activity exploration have resulted in the production of peptide analogs that are antagonists, one of which is clinically used (the B2R antagonist icatibant), and also non-peptide ligands for both receptor subtypes. The modification of kinin receptor ligands has made them resistant to extracellular or endosomal peptidases and/or produced bifunctional ligands, defined as agonist or antagonist peptide ligands conjugated with a chemical fluorophore (emitting in the whole spectrum, from the infrared to the ultraviolet), a drug-like moiety, an epitope, an isotope chelator/carrier, a cleavable sequence (thus forming a pro-drug) and even a fused protein. Dual molecular targets for specific modified peptides may be a source of side effects or of medically exploitable benefits. Biotechnological protein ligands for either receptor subtype have been produced: they are enhanced green fluorescent protein or the engineered peroxidase APEX2 fused to an agonist kinin sequence at their C-terminal terminus. Antibodies endowed with pharmacological actions (agonist, antagonist) at B2R have been reported, though not monoclonal antibodies. These findings define classes of alternative ligands of the kinin receptor of potential therapeutic and diagnostic value.
Collapse
Affiliation(s)
| | | | | | | | - Caroline Roy
- CHU de Québec - Université Laval, Québec, QC, G1 V 4G2, Canada
| | | | - Lajos Gera
- Department of Biochemistry, University of Colorado Denver, Aurora, CO, 80045, USA
| | | |
Collapse
|
32
|
Cardiovascular and Diabetic Medications That Cause Bradykinin-Mediated Angioedema. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2018; 5:610-615. [PMID: 28483314 DOI: 10.1016/j.jaip.2017.03.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 02/18/2017] [Accepted: 03/21/2017] [Indexed: 01/14/2023]
Abstract
Medication-induced angioedema is a bradykinin-mediated process that results from increased production or decreased degradation of bradykinin. These reactions are documented for several cardiac medications including blockers of the renin-angiotensin-aldosterone system (RAAS). Other cardiovascular and diabetes medications further increase the risk of medication-induced angioedema, particularly with concomitant use of RAAS inhibitors. Dipeptidyl peptidase IV inhibitors are a class of oral diabetic agents that affect bradykinin and substance P degradation and therefore can lead to angioedema. Neprilysin inhibitors are a separate class of cardiac medications, which includes sacubitril, and can lead to drug-induced angioedema especially when used in combination with RAAS inhibitors. This article discusses the proposed mechanisms by which these medications cause angioedema and how medication-induced angioedema differs from mast cell-mediated angioedema. It also details how to recognize medication-induced angioedema and the treatment options available.
Collapse
|
33
|
|
34
|
Levy RF, Serra AJ, Antonio EL, Dos Santos L, Bocalini DS, Pesquero JB, Bader M, Merino VF, de Oliveira HA, de Arruda Veiga EC, Silva JA, Tucci PJ. Cardiac morphofunctional characteristics of transgenic rats with overexpression of the bradykinin B1 receptor in the endothelium. Physiol Res 2017; 66:925-932. [PMID: 28937259 DOI: 10.33549/physiolres.933596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Our aim was to evaluate whether endothelial overexpressing of the bradykinin B1 receptor could be associated with altered left ventricular and myocardial performance. Echocardiography and hemodynamic were employed to assess left ventricular morphology and function in Sprague Dawley transgenic rats overexpressing the endothelial bradykinin B1 receptor (Tie2B1 rats). The myocardial inotropism was evaluated on papillary muscles contracting in vitro. In Tie2B1 animals, an enlarged left ventricular cavity and lower fractional shortening coupled with a lower rate of pressure change values indicated depressed left ventricular performance. Papillary muscle mechanics revealed that both Tie2B1 and wild-type rat groups had the same contractile capacities under basal conditions; however, in transgenic animals, there was accentuated inotropism due to post-pause potentiation. Following treatment with the Arg(9)-BK agonist, Tie2B1 papillary muscles displayed a reduction in myocardial inotropism. Endothelial B1 receptor overexpression has expanded the LV cavity and worsened its function. There was an exacerbated response of papillary muscle in vitro to a prolonged resting pause, and the use of a B1 receptor agonist impairs myocardial inotropism.
Collapse
Affiliation(s)
- R F Levy
- Universidade Federal da Paraíba, Joao Pessoa, Paraíba, Brazil, Universidade Federal de Sao Paulo, Sao Paulo, SP, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Collagenous proteins from black-barred halfbeak skin as a source of gelatin and bioactive peptides. Food Hydrocoll 2017. [DOI: 10.1016/j.foodhyd.2017.03.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
36
|
Soares DDM, Santos DR, Rummel C, Ott D, Melo MCC, Roth J, Calixto JB, Souza GEP. The relevance of kalikrein-kinin system via activation of B 2 receptor in LPS-induced fever in rats. Neuropharmacology 2017; 126:84-96. [PMID: 28826826 DOI: 10.1016/j.neuropharm.2017.08.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 08/05/2017] [Accepted: 08/11/2017] [Indexed: 12/21/2022]
Abstract
PURPOSE This study evaluated the involvement of endogenous kallikrein-kinin system and the bradykinin (BK) B1 and B2 receptors on LPS- induced fever and the POA cells involved in this response. MATERIAL AND METHODS Male Wistar rats received either i.v. (1 mg/kg), i.c.v. (20 nmol) or i.h. (2 nmol) injections of icatibant (B2 receptor antagonist) 30 or 60 min, respectively, before the stimuli. DALBK (B1 receptor antagonist) was given either 15min before BK (i.c.v.) or 30 min before LPS (i.v.). Captopril (5 mg/kg, sc.,) was given 1 h prior LPS or BK. Concentrations of BK and total kininogenon CSF, plasma and tissue kallikrein were evaluated. Rectal temperatures (rT) were assessed by telethermometry. Ca++ signaling in POA cells was performed in rat pup brain tissue microcultures. RESULTS Icatibant reduced LPS fever while, captopril exacerbated that response, an effect abolished by icatibant. Icatibant (i.h.) reduced fever to BK (i.h.) but not that induced by LPS (i.v.). BK increased intracellular calcium concentration in neurons and astrocytes. LPS increased levels of bradykinin, tissue kallikrein and total kininogen. BK (i.c.v.) increased rT and decreased tail skin temperature. Captopril potentiated BK-induced fever an effect abolished by icatibant. DALBK reduced the fever induced by BK. BK (i.c.v.) increased the CSF PGE2concentration. Effect abolished by indomethacin (i.p.). CONCLUSIONS LPS activates endogenous kalikrein-kinin system leading to production of BK, which by acting on B2-receptors of POA cells causes prostaglandin synthesis that in turn produces fever. Thus, a kinin B2-receptor antagonist that enters into the brain could constitute a new and interesting strategy to treat fever.
Collapse
Affiliation(s)
- Denis de Melo Soares
- Department of Medicament, Faculty of Pharmacy of Federal University of Bahia, Laboratory of Pharmacology, Ribeirão Preto, SP, Brazil.
| | - Danielle R Santos
- Pharmacology, Department of Physic and Chemistry, Faculty of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Christoph Rummel
- Veterinary Physiology, Faculty of Veterinary Medicine, Justus-Liebig-University of Giessen, Germany
| | - Daniela Ott
- Veterinary Physiology, Faculty of Veterinary Medicine, Justus-Liebig-University of Giessen, Germany
| | - Míriam C C Melo
- Pharmacology, Department of Physic and Chemistry, Faculty of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Joachim Roth
- Veterinary Physiology, Faculty of Veterinary Medicine, Justus-Liebig-University of Giessen, Germany
| | - João B Calixto
- Center of Innovation and Preclinical Research, Florianópolis, SC, Brazil
| | - Glória E P Souza
- Pharmacology, Department of Physic and Chemistry, Faculty of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
37
|
Abdelhedi O, Nasri R, Jridi M, Mora L, Oseguera-Toledo ME, Aristoy MC, Amara IB, Toldrá F, Nasri M. In silico analysis and antihypertensive effect of ACE-inhibitory peptides from smooth-hound viscera protein hydrolysate: Enzyme-peptide interaction study using molecular docking simulation. Process Biochem 2017. [DOI: 10.1016/j.procbio.2017.04.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
38
|
Effects of combination therapy with candesartan and ramipril on hypertension and related complications. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-017-0339-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
39
|
Xie Z, Chen F, Li WA, Geng X, Li C, Meng X, Feng Y, Liu W, Yu F. A review of sleep disorders and melatonin. Neurol Res 2017; 39:559-565. [PMID: 28460563 DOI: 10.1080/01616412.2017.1315864] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Zizhen Xie
- Department of Neurology, Beijing Haidian Hospital, Beijing, China
| | - Fei Chen
- Department of Neurology, Beijing Haidian Hospital, Beijing, China
| | - William A. Li
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaokun Geng
- Department of Neurology, Beijing Luhe Hospital Capital Medical University, Beijing, China
| | - Changhong Li
- Department of Neurology, Beijing Haidian Hospital, Beijing, China
| | - Xiaomei Meng
- Department of Neurology, Beijing Haidian Hospital, Beijing, China
| | - Yan Feng
- Department of Neurology, Beijing Haidian Hospital, Beijing, China
| | - Wei Liu
- Department of Neurology, Beijing Haidian Hospital, Beijing, China
| | - Fengchun Yu
- Department of Neurology, Beijing Haidian Hospital, Beijing, China
| |
Collapse
|
40
|
Mikrut K, Kupsz J, Kozlik J, Krauss H, Pruszynska-Oszmałek E, Gibas-Dorna M. Angiotensin-converting enzyme inhibitors reduce oxidative stress intensity in hyperglicemic conditions in rats independently from bradykinin receptor inhibitors. Croat Med J 2017; 57:371-80. [PMID: 27586552 PMCID: PMC5048232 DOI: 10.3325/cmj.2016.57.371] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim To investigate whether bradykinin-independent antioxidative effects of angiotensin-converting enzyme inhibitors (ACEIs) exist in acute hyperglycemia. Methods Male Wistar rats were divided into the normoglycemic group (n = 40) and the hyperglycemic group (n = 40). Hyperglycemia was induced by a single intraperitoneal injection of streptozotocin (STZ, 65 mg/kg body weight) dissolved in 0.1 mol/L citrate buffer (pH 4.5) 72 hours before sacrifice. The normoglycemic group received the same volume of citrate buffer. Each group was divided into five subgroups (n = 8): control group, captopril group, captopril + bradykinin B1 and B2 receptor antagonists group, enalapril group, and enalapril + bradykinin B1 and B2 receptor antagonists group. Captopril, enalapril, B1 and B2 receptor antagonists, or 0.15 mol/L NaCl were given at 2 and 1 hour before sacrifice. Oxidative status was determined by measuring the concentration of malondialdehyde and H2O2, and the activity of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx). Results In STZ-induced hyperglycemic rats ACEIs significantly reduced H2O2 and MDA concentration, while they significantly enhanced SOD and GPx activity. The hyperglycemic group treated simultaneously with ACEIs and bradykinin B1 and B2 receptor antagonists showed a significant decrease in H2O2 concentration compared to the control hyperglycemic group. Conclusion These results suggest the existence of additional antioxidative effect of ACEIs in hyperglycemic conditions, which is not related to the bradykinin mediation and the structure of the drug molecule.
Collapse
Affiliation(s)
- Kinga Mikrut
- Kinga Mikrut, Department of Physiology, Poznan University of Medical Sciences, Swiecickiego St., 6, 60-781 Poznan, Poland,
| | | | | | | | | | | |
Collapse
|
41
|
Oliveira-Paula GH, Luizon MR, Lacchini R, Fontana V, Silva PS, Biagi C, Tanus-Santos JE. Gene-Gene Interactions Among PRKCA, NOS3 and BDKRB2 Polymorphisms Affect the Antihypertensive Effects of Enalapril. Basic Clin Pharmacol Toxicol 2016; 120:284-291. [PMID: 27696692 DOI: 10.1111/bcpt.12682] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/22/2016] [Indexed: 01/08/2023]
Abstract
Protein kinase C (PKC) signalling is critically involved in the control of blood pressure. Angiotensin-converting enzyme inhibitors (ACEi) affect PKC expression and activity, which are partially associated with the responses to ACEi. We examined whether PRKCA (protein kinase C, alpha) polymorphisms (rs887797 C>T, rs1010544 T>C and rs16960228 G>A), or haplotypes, and gene-gene interactions within the ACEi pathway affect the antihypertensive responses in 104 hypertensive patients treated with enalapril as monotherapy. Patients were classified as poor responders (PR) or good responders (GR) to enalapril if their changes in mean arterial pressure were lower or higher than the median value, respectively. Multi-factor dimensionality reduction was used to characterize interactions among PRKCA, NOS3 (nitric oxide synthase 3) and BDKRB2 (bradykinin receptor B2) polymorphisms. The TC+CC genotypes for the rs1010544 polymorphism were more frequent in GR than in PR (p = 0.037). Conversely, the GA+AA genotypes for the rs16960228 polymorphism, and the CTA haplotype, were more frequent in PR than in GR (p = 0.040 and p = 0.008, respectively). Moreover, the GG genotype for the PRKCA rs16960228 polymorphism was associated with PR or GR depending on the genotypes for the rs2070744 (NOS3) and rs1799722 (BDKRB2) polymorphisms (p = 0.012). Our results suggest that PRKCA polymorphisms and gene-gene interactions within the ACEi pathway affect the antihypertensive responses to enalapril.
Collapse
Affiliation(s)
- Gustavo H Oliveira-Paula
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Marcelo R Luizon
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Riccardo Lacchini
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Vanessa Fontana
- Department of Pharmacology, State University of Campinas, Campinas, SP, Brazil
| | - Pamela S Silva
- Department of Pharmacology, State University of Campinas, Campinas, SP, Brazil
| | - Celso Biagi
- Santa Casa of Araçatuba, Araçatuba, SP, Brazil
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| |
Collapse
|
42
|
Dézsi CA, Szentes V. Effects of Angiotensin-Converting Enzyme Inhibitors and Angiotensin Receptor Blockers on Prothrombotic Processes and Myocardial Infarction Risk. Am J Cardiovasc Drugs 2016; 16:399-406. [PMID: 27580998 PMCID: PMC5126203 DOI: 10.1007/s40256-016-0185-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Acute ischemic events occur most frequently at dawn and in the early hours of the morning. The development of these severe clinical events exhibits a temporal relationship with changes in various hemodynamic, prothrombotic, and hormonal processes. The authors highlight not only these relationships but also the potential protective effect of increased bradykinin levels and the inhibition of different angiotensin II (AT-II) receptors (AT2, AT4) against unfavorable prothrombotic influences, which—based on studies to date—decreases the risk of acute cardiovascular events. Comparisons are presented between the different effects of angiotensin-converting enzyme inhibitors and angiotensin receptor blockers on factors that influence thrombus formation and myocardial infarction risk.
Collapse
Affiliation(s)
- Csaba András Dézsi
- Department of Cardiology, Petz Aladár County Teaching Hospital, Vasvári Pál str. 2-4, Gyor, 9024, Hungary.
| | - Veronika Szentes
- Department of Cardiology, Petz Aladár County Teaching Hospital, Vasvári Pál str. 2-4, Gyor, 9024, Hungary
| |
Collapse
|
43
|
Potentiation of Paclitaxel-Induced Pain Syndrome in Mice by Angiotensin I Converting Enzyme Inhibition and Involvement of Kinins. Mol Neurobiol 2016; 54:7824-7837. [PMID: 27844290 DOI: 10.1007/s12035-016-0275-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 10/30/2016] [Indexed: 10/20/2022]
Abstract
Paclitaxel is a chemotherapeutic agent used to treat solid tumours. However, it causes an acute and neuropathic pain syndrome that limits its use. Among the mechanisms involved in neuropathic pain caused by paclitaxel is activation of kinin receptors. Angiotensin converting enzyme (ACE) inhibitors can enhance kinin receptor signalling. The goal of this study was to evaluate the role of kinins on paclitaxel-associated acute pain syndromes (P-APS) and the effect of ACE inhibition on P-APS and paclitaxel-associated chronic peripheral neuropathy (P-CPN) in mice. Herein, we show that paclitaxel caused mechanical allodynia and spontaneous nociceptive behaviour that was reduced by antagonists of kinin receptors B1 (DALBk and SSR240612) and B2 (Hoe140 and FR173657). Moreover, enalapril (an ACE inhibitor) enhanced the mechanical allodynia induced by a low dose of paclitaxel. Likewise, paclitaxel injection inhibited ACE activity and increased the expressions of B1 and B2 receptors and bradykinin-related peptides levels in peripheral tissue. Together, our data support the involvement of kinin receptors in the P-APS and suggest kinin receptor antagonists to treat this syndrome. Because hypertension is the most frequent comorbidity affecting cancer patients, treatment of hypertension with ACE inhibitors in patients undergoing paclitaxel chemotherapy should be reviewed, since this could enhance the P-APS and P-CPN.
Collapse
|
44
|
First Report of Eurycoma longifolia Jack Root Extract Causing Relaxation of Aortic Rings in Rats. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1361508. [PMID: 27800486 PMCID: PMC5075299 DOI: 10.1155/2016/1361508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/12/2016] [Accepted: 09/14/2016] [Indexed: 12/29/2022]
Abstract
Although Eurycoma longifolia has been studied for erectile function, the blood pressure- (BP-) lowering effect has yet to be verified. Hence, this study aims at investigating the BP-lowering properties of the plant with a view to develop an antihypertensive agent that could also preserve erectile function. Ethanolic root extract was partitioned by hexane, dichloromethane (DCM), ethyl acetate, butanol, and water. The DCM fraction, found to be potent in relaxing phenylephrine- (PE-) precontracted rat aortic rings, was further purified by column chromatography. Subfraction DCM-II, being the most active in relaxing aortae, was studied for effects on the renin-angiotensin and kallikrein-kinin systems in aortic rings. The effect of DCM-II on angiotensin-converting enzyme (ACE) activity was also evaluated in vitro. Results showed that DCM-II reduced (p < 0.05) the contractions evoked by angiotensin I and angiotensin II (Ang II). In PE-precontracted rings treated with DCM-II, the Ang II-induced contraction was attenuated (p < 0.05) while bradykinin- (BK-) induced relaxation enhanced (p < 0.001). In vitro, DCM-II inhibited (p < 0.001) the activity of ACE. These data demonstrate that the vasodilatory effect of DCM-II appears to be mediated via inhibition of Ang II type 1 receptor and ACE as well as enhancement of Ang II type 2 receptor activation and BK activity.
Collapse
|
45
|
Abstract
Historically, the first described effect of an angiotensin converting enzyme (ACE) inhibitor was an increased activity of bradykinin, one of the substrates of ACE. However, in the subsequent years, molecular models describing the mechanism of action of ACE inhibitors in decreasing blood pressure and cardiovascular risk have focused mostly on the renin-angiotensin system. Nonetheless, over the last 20 years, the importance of bradykinin in regulating vasodilation, natriuresis, oxidative stress, fibrinolysis, inflammation, and apoptosis has become clearer. The affinity of ACE appears to be higher for bradykinin than for angiotensin I, thereby suggesting that ACE inhibitors may be more effective inhibitors of bradykinin degradation than of angiotensin II production. Data describing the effect of ACE inhibition on bradykinin signaling support the hypothesis that the most cardioprotective benefits attributed to ACE inhibition may be due to increased bradykinin signaling rather than to decreased angiotensin II signaling, especially when high dosages of ACE inhibitors are considered. In particular, modulation of bradykinin in the endothelium appears to be a major target of ACE inhibition. These new mechanistic concepts may lead to further development of strategies enhancing the bradykinin signaling.
Collapse
Affiliation(s)
- Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy.
| | - L Bortolotto
- Heart Institute-Hypertension Unit, Medical School University of Sao Paulo, São Paulo, Brazil
| |
Collapse
|
46
|
Raffai G, Lombard JH. Angiotensin-(1-7) Selectively Induces Relaxation and Modulates Endothelium-Dependent Dilation in Mesenteric Arteries of Salt-Fed Rats. J Vasc Res 2016; 53:105-118. [PMID: 27676088 DOI: 10.1159/000448714] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/26/2016] [Indexed: 12/19/2022] Open
Abstract
This study investigated the acute effects of angiotensin-(1-7) and AVE0991 on active tone and vasodilator responses to bradykinin and acetylcholine in isolated mesenteric arteries from Sprague-Dawley rats fed a high-salt (HS; 4% NaCl) versus a normal salt (NS; 0.4% NaCl) diet. Angiotensin-(1-7) and AVE0991 elicited relaxation, and angiotensin-(1-7) unmasked vasodilator responses to bradykinin in arteries from HS-fed rats. These effects of angiotensin-(1-7) and AVE0991 were inhibited by endothelium removal, A779, PD123319, HOE140 and L-NAME. Angiotensin-(1-7) also restored the acetylcholine-induced relaxation that was suppressed by the HS diet. Vasodilator responses to bradykinin and acetylcholine in the presence of angiotensin-(1-7) were mimicked by captopril and the AT2 receptor agonist CGP42112 in arteries from HS-fed rats. Thus, in contrast to salt-induced impairment of vascular relaxation in response to vasodilator stimuli, angiotensin-(1-7) induces endothelium-dependent and NO-mediated relaxation, unmasks bradykinin responses via activation of mas and AT2 receptors, and restores acetylcholine-induced vasodilation in HS-fed rats. AT2 receptor activation and angiotensin-converting enzyme (ACE) inhibition shared the ability of angiotensin-(1-7) to enhance bradykinin and acetylcholine responses in HS-fed rats. These findings suggest a therapeutic potential for mas and/or AT2 receptor activation and ACE inhibition in restoring endothelial function impaired by elevated dietary salt intake or other pathological conditions.
Collapse
Affiliation(s)
- Gábor Raffai
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wis., USA
| | | |
Collapse
|
47
|
Lin X, Bernloehr C, Hildebrandt T, Stadler FJ, Doods H, Wu D. Kinin B1 receptor blockade and ACE inhibition attenuate cardiac postinfarction remodeling and heart failure in rats. Toxicol Appl Pharmacol 2016; 305:153-160. [PMID: 27288733 DOI: 10.1016/j.taap.2016.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 05/18/2016] [Accepted: 06/06/2016] [Indexed: 10/21/2022]
Abstract
INTRODUCTION The aim of the present study was to evaluate the effects of the novel kinin B1 receptor antagonist BI113823 on postinfarction cardiac remodeling and heart failure, and to determine whether B1 receptor blockade alters the cardiovascular effects of an angiotensin 1 converting enzyme (ACE) inhibitor in rats. METHODS AND RESULTS Sprague Dawley rats were subjected to permanent occlusion of the left coronary artery. Cardiovascular function was determined at 6weeks postinfarction. Treatment with either B1 receptor antagonist (BI113823) or an ACE inhibitor (lisinopril) alone or in combination significantly reduced the heart weight-to-body weight and lung weight-to-body weight ratios, and improved postinfarction cardiac function as evidenced by greater cardiac output, the maximum rate of left ventricular pressure rise (±dP/dtmax), left ventricle ejection fraction, fractional shorting, better wall motion, and attenuation of elevated left ventricular end diastolic pressure (LVEDP). Furthermore, all three treatment groups exhibited significant reduction in cardiac interstitial fibrosis, collagen deposition, CD68 positive macrophages, neutrophils, and proinflammatory cytokine production (TNF-α and IL-1β), compared to vehicle controls. CONCLUSION The present study shows that treatment with the novel kinin B1 receptor antagonist, BI113823, reduces postinfarction cardiac remodeling and heart failure, and does not influence the cardiovascular effects of the ACE inhibitor.
Collapse
Affiliation(s)
- Xinchun Lin
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
| | | | | | - Florian J Stadler
- Shenzhen Engineering Laboratory for Advanced Technology of Ceramics, Shenzhen 518060, PR China.
| | - Henri Doods
- Boehringer Ingelheim Pharma GmbH & Co.KG, Biberach, Germany
| | - Dongmei Wu
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; Department of BIN Convergence Technology, Chonbuk National University, South Korea.
| |
Collapse
|
48
|
Seth MK, Hussain ME, Pasha S, Fahim M. Effects of a novel ACE inhibitor, 3-(3-thienyl)-l-alanyl-ornithyl-proline, on endothelial vasodilation and hepatotoxicity in l-NAME-induced hypertensive rats. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:1533-42. [PMID: 27143859 PMCID: PMC4844316 DOI: 10.2147/dddt.s77761] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Nitric oxide (NO) is a widespread biological mediator involved in many physiological and pathological processes, eg, in the regulation of vascular tone and hypertension. Chronic inhibition of NO synthase by NG-nitro-l-arginine methyl ester (l-NAME) hydrochloride results in the development of hypertension accompanied by an increase in vascular responsiveness to adrenergic stimuli. Recently, we developed a novel sulfur-containing angiotensin-converting enzyme inhibitor: 3-(3-thienyl)-l-alanyl-ornithyl-proline (TOP). Our previous studies indicated a superior nature of the molecule as an antihypertensive agent in spontaneously hypertensive rats (showing the involvement of renin–angiotensin–aldosterone system) in comparison to captopril. The aim of the present study was to investigate the effect of TOP on NO pathway in l-NAME-induced hypertensive rats, and captopril was included as the standard treatment group. Treatment with both TOP (20 mg/kg) and captopril (40 mg/kg) prevented the development of hypertension in l-NAME model, but TOP showed better restoration of NO and normal levels of angiotensin-converting enzyme. In addition, in vitro vasorelaxation assay showed an improvement in endothelium-dependent vasodilation in both the cases. Further, the biochemical (malondialdehyde, alanine aminotransferase, and aspartate aminotransferase) and the histopathological effects of TOP on rat liver tissues revealed a protective nature of TOP in comparison to captopril in the l-NAME model. In conclusion, TOP at 50% lesser dose than captopril was found to be better in the l-NAME model.
Collapse
Affiliation(s)
- Mahesh Kumar Seth
- Peptide Synthesis Laboratory, CSIR, Institute of Genomics and Integrative Biology, Delhi, India; Centre for Physiotherapy and Rehabilitation Sciences, Jamia Millia Islamia, New Delhi, India; Department of Physiology, Jamia Hamdard Deemed University, New Delhi, India
| | - M Ejaz Hussain
- Centre for Physiotherapy and Rehabilitation Sciences, Jamia Millia Islamia, New Delhi, India
| | - Santosh Pasha
- Peptide Synthesis Laboratory, CSIR, Institute of Genomics and Integrative Biology, Delhi, India
| | - Mohammad Fahim
- Department of Physiology, Jamia Hamdard Deemed University, New Delhi, India
| |
Collapse
|
49
|
Bas M, Greve J, Strassen U, Khosravani F, Hoffmann TK, Kojda G. Angioedema induced by cardiovascular drugs: new players join old friends. Allergy 2015; 70:1196-200. [PMID: 26119220 DOI: 10.1111/all.12680] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2015] [Indexed: 12/11/2022]
Abstract
During the last years, two new cardiovascular drug classes, namely inhibitors of DPP IV or neprilysin, have been developed. In both cases, there is clinical evidence for their potential to induce angioedema as known already from blockers of the renin-angiotensin-aldosterone system (RAAS). The majority of angioedema induced by DPP IV inhibitors occurs during concomitant treatment with ACEi and is therefore likely mediated by overactivation of bradykinin type 2 receptors (B2). In striking contrast, the molecular pathways causing angioedema induced by neprilysin inhibitors, that is, sacubitril, are unclear, although a contribution of bradykinin appears likely. Nevertheless, there is no clinical evidence suggesting that inhibition of B2 might relieve the symptoms and/or prevent invasive treatment including coniotomy or tracheotomy in angioedema caused by these drugs. Therefore, the risk of angioedema should always be considered, especially in ambulatory care situations where patients have no rapid access to intensive care.
Collapse
Affiliation(s)
- M. Bas
- Otorhinolaryngology Department; University Hospital Rechts der Isar; Munich Technical University; Munich Germany
| | - J. Greve
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery; Ulm University Medical Center; Ulm Germany
| | - U. Strassen
- Otorhinolaryngology Department; University Hospital Rechts der Isar; Munich Technical University; Munich Germany
| | - F. Khosravani
- Institute of Pharmacology and Clinical Pharmacology; Heinrich Heine University; Düsseldorf Germany
| | - T. K. Hoffmann
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery; Ulm University Medical Center; Ulm Germany
| | - G. Kojda
- Institute of Pharmacology and Clinical Pharmacology; Heinrich Heine University; Düsseldorf Germany
| |
Collapse
|
50
|
Role of Mas Receptor Antagonist A799 in Renal Blood Flow Response to Ang 1-7 after Bradykinin Administration in Ovariectomized Estradiol-Treated Rats. Adv Pharmacol Sci 2015; 2015:801053. [PMID: 26421009 PMCID: PMC4573425 DOI: 10.1155/2015/801053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/11/2015] [Accepted: 08/12/2015] [Indexed: 01/09/2023] Open
Abstract
Background. The accompanied role of Mas receptor (MasR), bradykinin (BK), and female sex hormone on renal blood flow (RBF) response to angiotensin 1-7 is not well defined. We investigated the role of MasR antagonist (A779) and BK on RBF response to Ang 1-7 infusion in ovariectomized estradiol-treated rats. Methods. Ovariectomized Wistar rats received estradiol (OVE) or vehicle (OV) for two weeks. Catheterized animals were subjected to BK and A799 infusion and mean arterial pressure (MAP), RBF, and renal vascular resistance (RVR) responses to Ang 1-7 (0, 100, and 300 ng kg−1 min−1) were determined. Results. Percentage change of RBF (%RBF) in response to Ang1-7 infusion increased in a dose-dependent manner. In the presence of BK, when MasR was not blocked, %RBF response to Ang 1-7 in OVE group was greater than OV group significantly (P < 0.05). Infusion of 300 ng kg−1 min−1 Ang 1-7 increased RBF by 6.9 ± 1.9% in OVE group versus 0.9 ± 1.8% in OV group. However when MasR was blocked, %RBF response to Ang 1-7 in OV group was greater than OVE group insignificantly. Conclusion. Coadministration of BK and A779 compared to BK alone increased RBF response to Ang 1-7 in vehicle treated rats. Such observation was not seen in estradiol treated rats.
Collapse
|