201
|
Hostrup A, Christensen GL, Bentzen BH, Liang B, Aplin M, Grunnet M, Hansen JL, Jespersen T. Functionally Selective AT1Receptor Activation Reduces Ischemia Reperfusion Injury. Cell Physiol Biochem 2012; 30:642-52. [DOI: 10.1159/000341445] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2012] [Indexed: 01/07/2023] Open
|
202
|
Chen DY, Lee Y, Van Tine BA, Searleman AC, Westergard TD, Liu H, Tu HC, Takeda S, Dong Y, Piwnica-Worms DR, Oh KJ, Korsmeyer SJ, Hermone A, Gussio R, Shoemaker RH, Cheng EHY, Hsieh JJD. A pharmacologic inhibitor of the protease Taspase1 effectively inhibits breast and brain tumor growth. Cancer Res 2011; 72:736-46. [PMID: 22166309 DOI: 10.1158/0008-5472.can-11-2584] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The threonine endopeptidase Taspase1 has a critical role in cancer cell proliferation and apoptosis. In this study, we developed and evaluated small molecule inhibitors of Taspase1 as a new candidate class of therapeutic modalities. Genetic deletion of Taspase1 in the mouse produced no overt deficiencies, suggesting the possibility of a wide therapeutic index for use of Taspase1 inhibitors in cancers. We defined the peptidyl motifs recognized by Taspase1 and conducted a cell-based dual-fluorescent proteolytic screen of the National Cancer Institute diversity library to identify Taspase1 inhibitors (TASPIN). On the basis of secondary and tertiary screens the 4-[(4-arsonophenyl)methyl]phenyl] arsonic acid NSC48300 was determined to be the most specific active compound. Structure-activity relationship studies indicated a crucial role for the arsenic acid moiety in mediating Taspase1 inhibition. Additional fluorescence resonance energy transfer-based kinetic analysis characterized NSC48300 as a reversible, noncompetitive inhibitor of Taspase1 (K(i) = 4.22 μmol/L). In the MMTV-neu mouse model of breast cancer and the U251 xenograft model of brain cancer, NSC48300 produced effective tumor growth inhibition. Our results offer an initial preclinical proof-of-concept to develop TASPINs for cancer therapy.
Collapse
Affiliation(s)
- David Y Chen
- Department of Medicine, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Pochynyuk O, Stockand JD. Role of Collecting Duct AT1a Receptors in Concentrating Urine. J Am Soc Nephrol 2011; 22:2144-5. [DOI: 10.1681/asn.2011100960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
204
|
Chen A, Aspiotis R, Campeau LC, Cauchon E, Chefson A, Ducharme Y, Falgueyret JP, Gagné S, Han Y, Houle R, Laliberté S, Larouche G, Lévesque JF, McKay D, Percival D. Renin inhibitors for the treatment of hypertension: Design and optimization of a novel series of spirocyclic piperidines. Bioorg Med Chem Lett 2011; 21:7399-404. [DOI: 10.1016/j.bmcl.2011.10.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 09/30/2011] [Accepted: 10/04/2011] [Indexed: 11/29/2022]
|
205
|
Safari F, Hajizadeh S, Shekarforoush S, Bayat G, Foadoddini M, Khoshbaten A. Influence of ramiprilat and losartan on ischemia reperfusion injury in rat hearts. J Renin Angiotensin Aldosterone Syst 2011; 13:29-35. [DOI: 10.1177/1470320311426025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Hypothesis/introduction: Our aim was to investigate whether a non-hypotensive dose of ramiprilat and losartan has myocardial protective effects during myocardial ischemia/reperfusion in vivo. Materials and methods: Three groups of rats were given 10 mg/kg per day of losartan for one (L-1W), four (L-4W) or 10 (L-10W) weeks. Another three groups were given 50 µg/kg per day of ramiprilat for one (R-1W), four (R-4W) or 10 (R-10W) weeks. The animals underwent 30 min of left anterior descending artery occlusion and subsequent reperfusion for 120 min. Results: Myocardial infarct size (IS) was reduced in R-1W (28.4 ± 6.3%, p < 0.001), R-4W (27.8 ± 7.4, p < 0.001), L-4W (31.8 ± 6%, p < 0.05) and L-10W (25.3 ± 5.7, p < 0.001) groups compared with a saline group (48.3 ± 7.8%). A significant reduction in the number of ventricular ectopic beats (VEBs) was noted in groups R-1W (209 ± 41, p < 0.01), R-4W (176 ± 39, p < 0.01), L-4W (215 ± 52, p < 0.05) and L-10W (191 ± 61, p < 0.01 vs. saline 329 ± 48). The incidence of irreversible ventricular fibrillation (VF) and mortality were decreased significantly only in L-10W group. There were no significant decreases in episodes of VT, the incidence of irreversible VF and mortality in all of the groups treated with ramiprilat. Conclusion: These data indicate that losartan and ramiprilat protect the heart against ischemia/reperfusion injury independently of their hemodynamic effects but in a time-dependent manner.
Collapse
Affiliation(s)
- Fatemeh Safari
- Department of Physiology, Tarbiat Modares University, Tehran, Iran
- Deptartment of Physiology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Sohrab Hajizadeh
- Department of Physiology, Tarbiat Modares University, Tehran, Iran
| | | | - Gholamreza Bayat
- Department of Physiology, Tarbiat Modares University, Tehran, Iran
| | - Mohsen Foadoddini
- Department of Physiology and Pharmacology, Birjand University of Medical Sciences, Birjand, Iran
| | - Ali Khoshbaten
- Exercise Physiology Research Center, Baqiatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
206
|
Jia L, Simpson RD, Yuan J, Xu Z, Zhao W, Cacatian S, Tice CM, Guo J, Ishchenko A, Singh SB, Wu Z, McKeever BM, Bukhtiyarov Y, Johnson JA, Doe CP, Harrison RK, McGeehan GM, Dillard LW, Baldwin JJ, Claremon DA. Discovery of VTP-27999, an Alkyl Amine Renin Inhibitor with Potential for Clinical Utility. ACS Med Chem Lett 2011; 2:747-51. [PMID: 24900262 DOI: 10.1021/ml200137x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 08/09/2011] [Indexed: 01/17/2023] Open
Abstract
Structure guided optimization of a series of nonpeptidic alkyl amine renin inhibitors allowed the rational incorporation of additional polar functionality. Replacement of the cyclohexylmethyl group occupying the S1 pocket with a (R)-(tetrahydropyran-3-yl)methyl group and utilization of a different attachment point led to the identification of clinical candidate 9. This compound demonstrated excellent selectivity over related and unrelated off-targets, >15% oral bioavailability in three species, oral efficacy in a double transgenic rat model of hypertension, and good exposure in humans.
Collapse
Affiliation(s)
- Lanqi Jia
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Robert D. Simpson
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Jing Yuan
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Zhenrong Xu
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Wei Zhao
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Salvacion Cacatian
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Colin M. Tice
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Joan Guo
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Alexey Ishchenko
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Suresh B. Singh
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Zhongren Wu
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Brian M. McKeever
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Yuri Bukhtiyarov
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Judith A. Johnson
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Christopher P. Doe
- GlaxoSmithKline, 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Richard K. Harrison
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Gerard M. McGeehan
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Lawrence W. Dillard
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - John J. Baldwin
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - David A. Claremon
- Vitae Pharmaceuticals, 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| |
Collapse
|
207
|
Li H, Prairie N, Udenigwe CC, Adebiyi AP, Tappia PS, Aukema HM, Jones PJH, Aluko RE. Blood pressure lowering effect of a pea protein hydrolysate in hypertensive rats and humans. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:9854-60. [PMID: 21854068 DOI: 10.1021/jf201911p] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The blood pressure lowering effect of a pea protein hydrolysate (PPH) that contained <3 kDa peptides, isolated by membrane ultrafiltration from the thermolysin digest of pea protein isolate (PPI), was examined using different rat models of hypertension as well as hypertensive human subjects. The PPH showed weak in vitro activities against renin and angiotensin converting enzyme (ACE) with inhibitory activities of 17 and 19%, respectively, at 1 mg/mL test concentration. Oral administration of the PPH to spontaneously hypertensive rats (SHR) at doses of 100 and 200 mg/kg body weight led to a lowering of hourly systolic blood pressure (SBP), with a maximum reduction of 19 mmHg at 4 h. In contrast, orally administered unhydrolyzed PPI had no blood pressure reducing effect in SHR, suggesting that thermolysin hydrolysis may have been responsible for releasing bioactive peptides from the native protein. Oral administration of the PPH to the Han:SPRD-cy rat (a model of chronic kidney disease) over an 8-week period led to 29 and 25 mmHg reductions in SBP and diastolic blood pressure, respectively. The PPH-fed rats had lower plasma levels of angiotensin II, the major vasopressor involved in development of hypertension, but there was no effect on plasma activity or renal mRNA levels of ACE. However, renal expression of renin mRNA levels was reduced by approximately 50% in the PPH-fed rats, suggesting that reduced renin may be responsible for the reduced levels of angiotensin II. In a 3-week randomized double blind placebo-controlled crossover human intervention trial (7 volunteers), significant (p<0.05) reductions (over placebo) in SBP of 5 and 6 mmHg were obtained in the second and third weeks, respectively, for the PPH group. Therefore, thermolysin derived bioactive peptides from PPH reduced blood pressure in hypertensive rats and human subjects, likely via effects on the renal angiotensin system.
Collapse
Affiliation(s)
- Huan Li
- Department of Human Nutritional Sciences and The Richardson Centre for Functional Foods and Nutraceuticals, University of Manitoba, Winnipeg, Manitoba, Canada R3T 2N2
| | | | | | | | | | | | | | | |
Collapse
|
208
|
Scheiper B, Matter H, Steinhagen H, Böcskei Z, Fleury V, McCort G. Structure-based optimization of potent 4- and 6-azaindole-3-carboxamides as renin inhibitors. Bioorg Med Chem Lett 2011; 21:5480-6. [PMID: 21840218 DOI: 10.1016/j.bmcl.2011.06.114] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 06/24/2011] [Accepted: 06/26/2011] [Indexed: 11/19/2022]
Affiliation(s)
- Bodo Scheiper
- Sanofi-Aventis, Deutschland GmbH, Chemical and Analytical Sciences, Building G878, D-65926 Frankfurt, Germany
| | | | | | | | | | | |
Collapse
|
209
|
Matter H, Scheiper B, Steinhagen H, Böcskei Z, Fleury V, McCort G. Structure-based design and optimization of potent renin inhibitors on 5- or 7-azaindole-scaffolds. Bioorg Med Chem Lett 2011; 21:5487-92. [DOI: 10.1016/j.bmcl.2011.06.112] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 06/24/2011] [Accepted: 06/26/2011] [Indexed: 10/18/2022]
|
210
|
Kantevari S, Addla D, Bagul PK, Sridhar B, Banerjee SK. Synthesis and evaluation of novel 2-butyl-4-chloro-1-methylimidazole embedded chalcones and pyrazoles as angiotensin converting enzyme (ACE) inhibitors. Bioorg Med Chem 2011; 19:4772-81. [DOI: 10.1016/j.bmc.2011.06.085] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2011] [Revised: 06/27/2011] [Accepted: 06/28/2011] [Indexed: 11/16/2022]
|
211
|
Esposito F, Galfetti M, Lava SAG, Balestra B, Bianchetti MG. Fetopathy probably associated to self-medication with a blocker of the renin-angiotensin system. Arch Gynecol Obstet 2011; 284:1321. [DOI: 10.1007/s00404-011-2027-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 07/14/2011] [Indexed: 10/18/2022]
|
212
|
Renin inhibitors for the treatment of hypertension: Design and optimization of a novel series of pyridone-substituted piperidines. Bioorg Med Chem Lett 2011; 21:3970-5. [DOI: 10.1016/j.bmcl.2011.05.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 05/03/2011] [Accepted: 05/04/2011] [Indexed: 11/21/2022]
|
213
|
Renin inhibitors for the treatment of hypertension: Design and optimization of a novel series of tertiary alcohol-bearing piperidines. Bioorg Med Chem Lett 2011; 21:3976-81. [DOI: 10.1016/j.bmcl.2011.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 05/03/2011] [Accepted: 05/04/2011] [Indexed: 11/17/2022]
|
214
|
Lévesque JF, Bleasby K, Chefson A, Chen A, Dubé D, Ducharme Y, Fournier PA, Gagné S, Gallant M, Grimm E, Hafey M, Han Y, Houle R, Lacombe P, Laliberté S, MacDonald D, Mackay B, Papp R, Tschirret-Guth R. Impact of passive permeability and gut efflux transport on the oral bioavailability of novel series of piperidine-based renin inhibitors in rodents. Bioorg Med Chem Lett 2011; 21:5547-51. [PMID: 21784634 DOI: 10.1016/j.bmcl.2011.06.095] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 06/17/2011] [Accepted: 06/20/2011] [Indexed: 12/31/2022]
Abstract
An oral bioavailability issue encountered during the course of lead optimization in the renin program is described herein. The low F(po) of pyridone analogs was shown to be caused by a combination of poor passive permeability and gut efflux transport. Substitution of pyridone ring for a more lipophilic moiety (logD>1.7) had minimal effect on rMdr1a transport but led to increased passive permeability (P(app)>10 × 10(-6) cm/s), which contributed to overwhelm gut transporters and increase rat F(po). LogD and in vitro passive permeability determination were found to be key in guiding SAR and improve oral exposure of renin inhibitors.
Collapse
Affiliation(s)
- Jean-François Lévesque
- Merck Frosst Centre for Therapeutic Research, 16711 Trans Canada Highway, Kirkland, Québec, Canada H9H 3L1.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
215
|
Campeau LC, Dolman SJ, Gauvreau D, Corley E, Liu J, Guidry EN, Ouellet SG, Steinhuebel D, Weisel M, O’Shea PD. Convergent Kilo-Scale Synthesis of a Potent Renin Inhibitor for the Treatment of Hypertension. Org Process Res Dev 2011. [DOI: 10.1021/op2001063] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Louis-Charles Campeau
- Global Process Chemistry, Merck-Frosst, 16711 Trans Canada Highway, Kirkland, Québec H9H 3L1, Canada
| | - Sarah J. Dolman
- Global Process Chemistry, Merck-Frosst, 16711 Trans Canada Highway, Kirkland, Québec H9H 3L1, Canada
| | - Danny Gauvreau
- Global Process Chemistry, Merck-Frosst, 16711 Trans Canada Highway, Kirkland, Québec H9H 3L1, Canada
| | - Ed Corley
- Global Process Chemistry, Merck, P.O. Box 2000, Rahway, New Jersey 07065, United States
| | - Jinchu Liu
- Global Process Chemistry, Merck, P.O. Box 2000, Rahway, New Jersey 07065, United States
| | - Erin N. Guidry
- Global Process Chemistry, Merck, P.O. Box 2000, Rahway, New Jersey 07065, United States
| | - Stéphane G. Ouellet
- Global Process Chemistry, Merck-Frosst, 16711 Trans Canada Highway, Kirkland, Québec H9H 3L1, Canada
| | - Dietrich Steinhuebel
- Global Process Chemistry, Merck, P.O. Box 2000, Rahway, New Jersey 07065, United States
| | - Mark Weisel
- Global Process Chemistry, Merck, P.O. Box 2000, Rahway, New Jersey 07065, United States
| | - Paul D. O’Shea
- Global Process Chemistry, Merck-Frosst, 16711 Trans Canada Highway, Kirkland, Québec H9H 3L1, Canada
| |
Collapse
|
216
|
Wu T, Zhou X, Deng Y, Jing Q, Li M, Yuan L. In vitro studies of Gynura divaricata (L.) DC extracts as inhibitors of key enzymes relevant for type 2 diabetes and hypertension. JOURNAL OF ETHNOPHARMACOLOGY 2011; 136:305-308. [PMID: 21570455 DOI: 10.1016/j.jep.2011.04.059] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 04/12/2011] [Accepted: 04/20/2011] [Indexed: 05/30/2023]
Abstract
AIM OF THE STUDY To evaluate traditionally used herb, Gynura divaricata (L.) DC (Bai Bei San Qi) as in vitro inhibitors of key enzymes involved in the pathogenesis of hyperglycemia and hypertension. We also determined the distribution of enzyme inhibitory activities in different aqueous and non-aqueous extracts. MATERIALS AND METHODS The water extract (extract 1) from the aerial parts of Gynura divaricata (L.) was prepared first and then partitioned sequentially with n-butanol, ethyl acetate, and macroporous adsorptive resin (HPD-40) to yield extracts 2-4; the remaining water phase was named extract 5. Angiotensin-1 converting enzyme (ACE), α-amylase α-glycosidase inhibitory activities of the extracts were determined in vitro and chemical composition including total sugar, protein, flavonoid and total alkaloids in the extract were also evaluated. RESULTS The water extract of this herb significantly inhibited (p<0.05) ACE activity (IC(50)=0.37 mg/ml) and showed a moderate potential hypoglycemic effect via in vitro α-amylase (IC(50)=1.36 mg/ml) and α-glycosidase (IC(50)=2.17 mg/ml) inhibition in dose-dependent manner. Further partitioning of the water extract (extracts 2-4) resulted in higher α-amylase inhibitory activities in extract 2 and 3. For α-glycosidase inhibition, extract 3 gave the highest inhibition. ACE inhibitory activities of the extracts were not improved by partitioning. Sugar, protein, flavonoid and alkaloid were found in water extract but only a small portion was partitioned in the n-butanol extract. However, a large portion of the flavonoids and alkaloids were found in ethyl acetate extract. CONCLUSION The results confirmed potential empirical use of Gynura divaricata (L.) DC for the management of hyperglycemia as well as hypertension. The active compounds for inhibition of α-amylase and α-glycosidase inhibition were flavonoids and alkaloids while ACE inhibition probably resulted from synergic effects of all the herb compounds.
Collapse
Affiliation(s)
- Tingting Wu
- Chemistry Institute of Pharmaceutical Resources, College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, People's Republic of China
| | | | | | | | | | | |
Collapse
|
217
|
|
218
|
Oudit GY, Penninger JM. Recombinant Human Angiotensin-Converting Enzyme 2 as a New Renin-Angiotensin System Peptidase for Heart Failure Therapy. Curr Heart Fail Rep 2011; 8:176-83. [DOI: 10.1007/s11897-011-0063-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
219
|
Guimarães PB, Alvarenga ÉC, Siqueira PD, Paredes-Gamero EJ, Sabatini RA, Morais RL, Reis RI, Santos EL, Teixeira LG, Casarini DE, Martin RP, Shimuta SI, Carmona AK, Nakaie CR, Jasiulionis MG, Ferreira AT, Pesquero JL, Oliveira SM, Bader M, Costa-Neto CM, Pesquero JB. Angiotensin II Binding to Angiotensin I–Converting Enzyme Triggers Calcium Signaling. Hypertension 2011; 57:965-72. [PMID: 21422380 DOI: 10.1161/hypertensionaha.110.167171] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Angiotensin (Ang) I–converting enzyme (ACE) is involved in the control of blood pressure by catalyzing the conversion of Ang I into the vasoconstrictor Ang II and degrading the vasodilator peptide bradykinin. Human ACE also functions as a signal transduction molecule, and the binding of ACE substrates or its inhibitors initiates a series of events. In this study, we examined whether Ang II could bind to ACE generating calcium signaling. Chinese hamster ovary cells transfected with an ACE expression vector reveal that Ang II is able to bind with high affinity to ACE in the absence of the Ang II type 1 and type 2 receptors and to activate intracellular signaling pathways, such as inositol 1,4,5-trisphosphate and calcium. These effects could be blocked by the ACE inhibitor, lisinopril. Calcium mobilization was specific for Ang II, because other ACE substrates or products, namely Ang 1-7, bradykinin, bradykinin 1-5, and
N
-acetyl-seryl-aspartyl-lysyl-proline, did not trigger this signaling pathway. Moreover, in Tm5, a mouse melanoma cell line endogenously expressing ACE but not Ang II type 1 or type 2 receptors, Ang II increased intracellular calcium and reactive oxygen species. In conclusion, we describe for the first time that Ang II can interact with ACE and evoke calcium and other signaling molecules in cells expressing only ACE. These findings uncover a new mechanism of Ang II action and have implications for the understanding of the renin-Ang system.
Collapse
Affiliation(s)
- Paola B. Guimarães
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Érika C. Alvarenga
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Paula D. Siqueira
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Edgar J. Paredes-Gamero
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Regiane A. Sabatini
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Rafael L.T. Morais
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Rosana I. Reis
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Edson L. Santos
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Luis G.D. Teixeira
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Dulce E. Casarini
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Renan P. Martin
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Suma I. Shimuta
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Adriana K. Carmona
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Clovis R. Nakaie
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Miriam G. Jasiulionis
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Alice T. Ferreira
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Jorge L. Pesquero
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Suzana M. Oliveira
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Michael Bader
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - Claudio M. Costa-Neto
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| | - João B. Pesquero
- From the Departamento de Biofísica (P.B.G., É.C.A., P.D.S., E.J.P.-G., R.A.S., R.L.T.M., L.G.D.T., R.P.M., S.I.S., A.K.C., C.R.N., A.T.F., S.M.O., J.B.P.), Departamento de Medicina (D.E.C.), and Departamento de Farmacologia (M.G.J.), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Bioquímica e Imunologia (R.I.R., C.M.C.-N.), Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Ciências Biológicas e Ambientais (E.L.S.), Universidade
| |
Collapse
|
220
|
Wang X, Wu S, Xu D, Xie D, Guo H. Inhibitor and substrate binding by angiotensin-converting enzyme: quantum mechanical/molecular mechanical molecular dynamics studies. J Chem Inf Model 2011; 51:1074-82. [PMID: 21520937 DOI: 10.1021/ci200083f] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Angiotensin-converting enzyme (ACE) is an important zinc-dependent hydrolase responsible for converting the inactive angiotensin I to the vasoconstrictor angiotensin II and for inactivating the vasodilator bradykinin. However, the substrate binding mode of ACE has not been completely understood. In this work, we propose a model for an ACE Michaelis complex based on two known X-ray structures of inhibitor-enzyme complexes. Specifically, the human testis angiotensin-converting enzyme (tACE) complexed with two clinic drugs were first investigated using a combined quantum mechanical and molecular mechanical (QM/MM) approach. The structural parameters obtained from the 550 ps molecular dynamics simulations are in excellent agreement with the X-ray structures, validating the QM/MM approach. Based on these structures, a model for the Michaelis complex was proposed and simulated using the same computational protocol. Implications to ACE catalysis are discussed.
Collapse
Affiliation(s)
- Xuemei Wang
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry, Sichuan University , Chengdu, Sichuan 610064, China
| | | | | | | | | |
Collapse
|
221
|
|
222
|
Aspiotis R, Chen A, Cauchon E, Dubé D, Falgueyret JP, Gagné S, Gallant M, Grimm EL, Houle R, Juteau H, Lacombe P, Laliberté S, Lévesque JF, MacDonald D, McKay D, Percival MD, Roy P, Soisson SM, Wu T. The discovery and synthesis of potent zwitterionic inhibitors of renin. Bioorg Med Chem Lett 2011; 21:2430-6. [DOI: 10.1016/j.bmcl.2011.02.067] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 02/15/2011] [Indexed: 11/26/2022]
|
223
|
Molinaro C, Shultz S, Roy A, Lau S, Trinh T, Angelaud R, O’Shea PD, Abele S, Cameron M, Corley E, Funel JA, Steinhuebel D, Weisel M, Krska S. A Practical Synthesis of Renin Inhibitor MK-1597 (ACT-178882) via Catalytic Enantioselective Hydrogenation and Epimerization of Piperidine Intermediate. J Org Chem 2011; 76:1062-71. [DOI: 10.1021/jo102070e] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Carmela Molinaro
- Department of Process Research, Merck, 16711 Autoroute Transcanadienne, Kirkland, Québec, Canada H9H 3L1
| | - Scott Shultz
- Department of Process Research, Merck, P.O. Box 2000, Rahway, New Jersey 07065, United States
| | - Amélie Roy
- Department of Process Research, Merck, 16711 Autoroute Transcanadienne, Kirkland, Québec, Canada H9H 3L1
| | - Stephen Lau
- Department of Process Research, Merck, 16711 Autoroute Transcanadienne, Kirkland, Québec, Canada H9H 3L1
| | - Thao Trinh
- Department of Process Research, Merck, 16711 Autoroute Transcanadienne, Kirkland, Québec, Canada H9H 3L1
| | - Rémy Angelaud
- Department of Process Research, Merck, 16711 Autoroute Transcanadienne, Kirkland, Québec, Canada H9H 3L1
| | - Paul D. O’Shea
- Department of Process Research, Merck, 16711 Autoroute Transcanadienne, Kirkland, Québec, Canada H9H 3L1
| | - Stefan Abele
- Department of Process Research Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, 4123 Allschwil, Switzerland
| | - Mark Cameron
- Department of Process Research, Merck, P.O. Box 2000, Rahway, New Jersey 07065, United States
| | - Ed Corley
- Department of Process Research, Merck, P.O. Box 2000, Rahway, New Jersey 07065, United States
| | - Jacques-Alexis Funel
- Department of Process Research Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, 4123 Allschwil, Switzerland
| | - Dietrich Steinhuebel
- Department of Process Research, Merck, P.O. Box 2000, Rahway, New Jersey 07065, United States
| | - Mark Weisel
- Department of Process Research, Merck, P.O. Box 2000, Rahway, New Jersey 07065, United States
| | - Shane Krska
- Department of Process Research, Merck, P.O. Box 2000, Rahway, New Jersey 07065, United States
| |
Collapse
|
224
|
The angiotensin II type 1 receptor antagonist Losartan binds and activates bradykinin B2 receptor signaling. ACTA ACUST UNITED AC 2010; 167:21-5. [PMID: 21115072 DOI: 10.1016/j.regpep.2010.11.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Revised: 10/05/2010] [Accepted: 11/16/2010] [Indexed: 11/20/2022]
Abstract
The angiotensin II type 1 receptor (AT1R) blocker (ARB) Losartan has cardioprotective effects during ischemia-reperfusion injury and inhibits reperfusion arrhythmias -effects that go beyond the benefits of lowering blood pressure. The renin-angiotensin and kallikrein-kinin systems are intricately connected and some of the cardioprotective effects of Losartan are abolished by blocking the bradykinin B2 receptor (B2R) signaling. In this study, we investigated the ability of six clinically available ARBs to specifically bind and activate the B2R. First, we investigated their ability to activate phosphoinositide (PI) hydrolysis in COS-7 cells transiently expressing the B2R. We found that only Losartan activated the B2R, working as a partial agonist compared to the endogenous ligand bradykinin. This effect was blocked by the B2R antagonist HOE 140. A competitive binding analysis revealed that Losartan does not significantly compete with bradykinin and does not change the binding affinity of bradykinin on the B2R. Furthermore, Losartan but not Candesartan mimicked the ability of bradykinin to increase the recovery of contractile force after metabolic stress in rat atrial tissue strips. In conclusion, Losartan is a partial agonist of the B2R through direct binding and activation, suggesting that B2R agonism could partly explain the beneficial effects of Losartan.
Collapse
|
225
|
Zhenfeng Zheng, Huilan Shi, Junya Jia, Dong Li, Shan Lin. A systematic review and meta-analysis of aliskiren and angiotension receptor blockers in the management of essential hypertension. J Renin Angiotensin Aldosterone Syst 2010; 12:102-12. [PMID: 21059822 DOI: 10.1177/1470320310381912] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Aliskiren is a novel antihypertensive agent and the first direct renin inhibitor (DRI) in clinical use. Several clinical trials have compared DRI with angiotensin receptor blockers (ARBs) in the management of essential hypertension. However, systematic comparison of efficacy and safety between DRIs and ARBs is still lacking. We reviewed randomized controlled trials (RCTs) comparing aliskiren with ARBs for net reduction of blood pressure from baseline, achieved rate of control, and incidences of common and serious adverse events. Weighted mean differences (WMD) and relative risk (RR) with 95% confidence intervals (CI) were calculated for continuous and dichotomous data, respectively. Seven RCTs with 5488 patients were included in this meta-analysis. We compared the efficacy of aliskiren and ARBs in reducing systolic blood pressure (SBP) and diastolic blood pressure (DBP). No differences were found between the two groups. Aliskiren combined with ARBs was superior to aliskiren monotherapy at the maximum recommended dose on SBP and DBP reduction. (WMD -4.80, 95% CI -6.22— -3.39, p < 0.0001; WMD -2.96, 95% CI -4.63—-1.28, p = 0.0001; respectively). Similar results were found with aliskiren combined with ARBs versus ARB monotherapy (WMD -4.43, 95% CI -5.91—-2.96, p < 0.0001; WMD -2.40; 95% CI -3.41—-1.39, p < 0.0001; respectively). No differences were found in adverse events between the aliskiren and ARB groups. Similar results were found with aliskiren and ARB combination therapy and its respective monotherapy. We conclude that aliskiren’s BP-lowering capabilities were comparable to those of ARBs. Aliskiren and ARB combination therapy provided more effective BP reduction than each respective monotherapy without increasing adverse events.
Collapse
Affiliation(s)
- Zhenfeng Zheng
- Department of Nephrology, The First Affiliated Hospital of Tianjin Medical University, China
| | - Huilan Shi
- Department of Radiology, The First Affiliated Hospital of Tianjin Medical University, China
| | - Junya Jia
- Department of Nephrology, The First Affiliated Hospital of Tianjin Medical University, China
| | - Dong Li
- Department of Nephrology, The First Affiliated Hospital of Tianjin Medical University, China
| | - Shan Lin
- Department of Nephrology, The First Affiliated Hospital of Tianjin Medical University, China
| |
Collapse
|
226
|
Discovery and optimization of a new class of potent and non-chiral indole-3-carboxamide-based renin inhibitors. Bioorg Med Chem Lett 2010; 20:6268-72. [DOI: 10.1016/j.bmcl.2010.08.092] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 08/16/2010] [Accepted: 08/17/2010] [Indexed: 11/15/2022]
|
227
|
Li L, Zhou N, Gong H, Wu J, Lin L, Komuro I, Ge J, Zou Y. Comparison of angiotensin II type 1-receptor blockers to regress pressure overload-induced cardiac hypertrophy in mice. Hypertens Res 2010; 33:1289-97. [DOI: 10.1038/hr.2010.182] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
228
|
da Silveira KD, Coelho FM, Vieira AT, Sachs D, Barroso LC, Costa VV, Bretas TLB, Bader M, de Sousa LP, da Silva TA, dos Santos RAS, Simões e Silva AC, Teixeira MM. Anti-inflammatory effects of the activation of the angiotensin-(1-7) receptor, MAS, in experimental models of arthritis. THE JOURNAL OF IMMUNOLOGY 2010; 185:5569-76. [PMID: 20935211 DOI: 10.4049/jimmunol.1000314] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Activation of the renin-angiotensin (Ang) system induces inflammation via interaction between Ang II and type 1 receptor on leukocytes. The relevance of the new arm of the renin-Ang system, namely Ang-converting enzyme-2/Ang-(1-7)/Mas receptor, for inflammatory responses is not known and was investigated in this study. For this purpose, two experimental models were used: Ag-induced arthritis (AIA) in mice and adjuvant-induced arthritis (AdIA) in rats. Male C57BL/6 wild-type or Mas(-/-) mice were subjected to AIA and treated with Ang-(1-7), the Mas agonist AVE 0991, or vehicle. AdIA was performed in female rats that were given AVE 0991 or vehicle. In wild-type mice, Mas protein is expressed in arthritic joints. Administration of AVE 0991 or Ang-(1-7) decreased AIA-induced neutrophil accumulation, hypernociception, and production of TNF-α, IL-1β, and CXCL1. Histopathological analysis showed significant reduction of inflammation. Mechanistically, AVE 0991 reduced leukocyte rolling and adhesion, even when given after Ag challenge. Mas(-/-) mice subjected to AIA developed slightly more pronounced inflammation, as observed by greater neutrophil accumulation and cytokine release. Administration of AVE 0991 was without effect in Mas(-/-) mice subjected to AIA. In rats, administration of AVE 0991 decreased edema, neutrophil accumulation, histopathological score, and production of IL-1β and CXCL1 induced by AdIA. Therefore, activation of Mas receptors decreases neutrophil influx and cytokine production and causes significant amelioration of arthritis in experimental models of arthritis in rats and mice. This approach might represent a novel therapeutic opportunity for arthritis.
Collapse
Affiliation(s)
- Kátia Daniela da Silveira
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
229
|
Zhong J, Basu R, Guo D, Chow FL, Byrns S, Schuster M, Loibner H, Wang XH, Penninger JM, Kassiri Z, Oudit GY. Angiotensin-converting enzyme 2 suppresses pathological hypertrophy, myocardial fibrosis, and cardiac dysfunction. Circulation 2010; 122:717-28, 18 p following 728. [PMID: 20679547 DOI: 10.1161/circulationaha.110.955369] [Citation(s) in RCA: 346] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Angiotensin-converting enzyme 2 (ACE2) is a pleiotropic monocarboxypeptidase capable of metabolizing several peptide substrates. We hypothesized that ACE2 is a negative regulator of angiotensin II (Ang II)-mediated signaling and its adverse effects on the cardiovascular system. METHODS AND RESULTS Ang II infusion (1.5 mg x kg(-1) x d(-1)) for 14 days resulted in worsening cardiac fibrosis and pathological hypertrophy in ACE2 knockout (Ace2(-/y)) mice compared with wild-type (WT) mice. Daily treatment of Ang II-infused wild-type mice with recombinant human ACE2 (rhACE2; 2 mg x kg(-1) x d(-1) IP) blunted the hypertrophic response and expression of hypertrophy markers and reduced Ang II-induced superoxide production. Ang II-mediated myocardial fibrosis and expression of procollagen type I alpha 1, procollagen type III alpha 1, transforming growth factor-beta1, and fibronectin were also suppressed by rhACE2. Ang II-induced diastolic dysfunction was inhibited by rhACE2 in association with reduced plasma and myocardial Ang II and increased plasma Ang 1-7 levels. rhACE2 treatment inhibited Ang II-mediated activation of protein kinase C-alpha and protein kinase C-beta1 protein levels and phosphorylation of the extracellular signal-regulated 1/2, Janus kinase 2, and signal transducer and activator of transcription 3 signaling pathways in wild-type mice. A subpressor dose of Ang II (0.15 mg . kg(-1) . d(-1)) resulted in a milder phenotype that was strikingly attenuated by rhACE2 (2 mg x kg(-1) x d(-1) IP). In adult ventricular cardiomyocytes and cardiofibroblasts, Ang II-mediated superoxide generation, collagen production, and extracellular signal-regulated 1/2 signaling were inhibited by rhACE2 in an Ang 1-7-dependent manner. Importantly, rhACE2 partially prevented the development of dilated cardiomyopathy in pressure-overloaded wild-type mice. CONCLUSIONS Elevated Ang II induced hypertension, myocardial hypertrophy, fibrosis, and diastolic dysfunction, which were exacerbated by ACE2 deficiency, whereas rhACE2 attenuated Ang II- and pressure-overload-induced adverse myocardial remodeling. Hence, ACE2 is an important negative regulator of Ang II-induced heart disease and suppresses adverse myocardial remodeling.
Collapse
Affiliation(s)
- JiuChang Zhong
- Division of Cardiology, Department of Medicine, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
230
|
Ragonesi PD. Effects of angiotensin II receptor blockers on insulin resistance. Hypertens Res 2010; 33:778. [DOI: 10.1038/hr.2010.100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
231
|
A small difference in the molecular structure of angiotensin II receptor blockers induces AT₁ receptor-dependent and -independent beneficial effects. Hypertens Res 2010; 33:1044-52. [PMID: 20668453 DOI: 10.1038/hr.2010.135] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Angiotensin II (Ang II) type 1 (AT₁) receptor blockers (ARBs) induce multiple pharmacological beneficial effects, but not all ARBs have the same effects and the molecular mechanisms underlying their actions are not certain. In this study, irbesartan and losartan were examined because of their different molecular structures (irbesartan has a cyclopentyl group whereas losartan has a chloride group). We analyzed the binding affinity and production of inositol phosphate (IP), monocyte chemoattractant protein-1 (MCP-1) and adiponectin. Compared with losartan, irbesartan showed a significantly higher binding affinity and slower dissociation rate from the AT₁ receptor and a significantly higher degree of inverse agonism and insurmountability toward IP production. These effects of irbesartan were not seen with the AT₁-Y113A mutant receptor. On the basis of the molecular modeling of the ARBs-AT₁ receptor complex and a mutagenesis study, the phenyl group at Tyr(113) in the AT₁ receptor and the cyclopentyl group of irbesartan may form a hydrophobic interaction that is stronger than the losartan-AT₁ receptor interaction. Interestingly, irbesartan inhibited MCP-1 production more strongly than losartan. This effect was mediated by the inhibition of nuclear factor-kappa B activation that was independent of the AT₁ receptor in the human coronary endothelial cells. In addition, irbesartan, but not losartan, induced significant adiponectin production that was mediated by peroxisome proliferator-activated receptor-γ activation in 3T3-L1 adipocytes, and this effect was not mediated by the AT₁ receptor. In conclusion, irbesartan induced greater beneficial effects than losartan due to small differences between their molecular structures, and these differential effects were both dependent on and independent of the AT₁ receptor.
Collapse
|
232
|
Hanessian S, Guesné S, Chénard E. Total synthesis of "aliskiren": the first Renin inhibitor in clinical practice for hypertension. Org Lett 2010; 12:1816-9. [PMID: 20235519 DOI: 10.1021/ol100427v] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We report a "macrocycle route" toward aliskiren, a drug presently marketed for the treatment of hypertension, using a highly stereocontrolled approach starting from a common "isopropyl chiron". Highlights of the synthesis include a challenging RCM reaction to produce a nine-membered unsaturated lactone, a highly stereoselective catalytic Du Bois aziridination, and a regio- and diastereoselective aziridine ring-opening to a vicinal amino alcohol.
Collapse
Affiliation(s)
- Stephen Hanessian
- Department of Chemistry, Universite de Montreal, C.P. 6128, Succ. Centre-Ville, Montreal, Quebec H3C 3J7, Canada.
| | | | | |
Collapse
|
233
|
Akif M, Ntai I, Sturrock ED, Isaac RE, Bachmann BO, Acharya KR. Crystal structure of a phosphonotripeptide K-26 in complex with angiotensin converting enzyme homologue (AnCE) from Drosophila melanogaster. Biochem Biophys Res Commun 2010; 398:532-6. [DOI: 10.1016/j.bbrc.2010.06.113] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Accepted: 06/27/2010] [Indexed: 10/19/2022]
|
234
|
Abstract
Despite the existence of established, effective therapies for hypertension, new methods of blood pressure and cardiovascular risk reduction are still needed. Novel approaches are targeted towards treating resistant hypertension, improving blood-pressure control, and achieving further risk reduction beyond blood-pressure lowering. Modulation of the renin-angiotensin-aldosterone system (RAAS) provides the rationale for current antihypertensive therapies, including the relatively new agents eplerenone and aliskiren. Novel targets for antihypertensive therapy are also likely to be RAAS-related. The stimulation of angiotensin II type 2 receptors, or supplementation with renalase, could counteract the effects of angiotensin II type 1 receptor stimulation or catecholamine release. Combined angiotensin-converting-enzyme and neutral endopeptidase blockade decreases blood pressure, but is associated with a high incidence of angioedema. Aldosterone synthase inhibitors might improve tolerability in aldosterone antagonism. A (pro)renin-receptor blocker could prevent the deleterious angiotensin-independent actions of renin that are not inhibited by aliskiren. Finally, new minimally invasive surgical procedures have revived the concept of renal denervation, and could be a therapeutic option for patients with resistant hypertension. All of these strategies are exciting prospects, but which of them will prove valuable in clinical setting remains to be discovered.
Collapse
Affiliation(s)
- Ludovit Paulis
- Center for Cardiovascular Research, Charité-Universitätsmedizin, Hessische Strasse 3-4, 10115 Berlin, Germany
| | | |
Collapse
|
235
|
Chen A, Bayly C, Bezençon O, Richard-Bildstein S, Dubé D, Dubé L, Gagné S, Gallant M, Gaudreault M, Grimm E, Houle R, Lacombe P, Laliberté S, Lévesque JF, Liu S, MacDonald D, Mackay B, Martin D, McKay D, Powell D, Remen L, Soisson S, Toulmond S. Design and optimization of a substituted amino propanamide series of renin inhibitors for the treatment of hypertension. Bioorg Med Chem Lett 2010; 20:2204-9. [PMID: 20206513 DOI: 10.1016/j.bmcl.2010.02.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Accepted: 02/08/2010] [Indexed: 11/25/2022]
Abstract
The discovery and SAR of a new series of substituted amino propanamide renin inhibitors are herein described. This work has led to the preparation of compounds with in vitro and in vivo profiles suitable for further development. Specifically, challenges pertaining to oral bioavailability, covalent binding and time-dependent CYP 3A4 inhibition were overcome thereby culminating in the identification of compound 50 as an optimized renin inhibitor with good efficacy in the hypertensive double-transgenic rat model.
Collapse
Affiliation(s)
- Austin Chen
- Merck Frosst Centre for Therapeutic Research, 16711 Trans Canada Highway, Kirkland, Québec, Canada H9H 3L1.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
236
|
Li NC, Lee A, Whitmer RA, Kivipelto M, Lawler E, Kazis LE, Wolozin B. Use of angiotensin receptor blockers and risk of dementia in a predominantly male population: prospective cohort analysis. BMJ 2010; 340:b5465. [PMID: 20068258 PMCID: PMC2806632 DOI: 10.1136/bmj.b5465] [Citation(s) in RCA: 343] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To investigate whether angiotensin receptor blockers protect against Alzheimer's disease and dementia or reduce the progression of both diseases. DESIGN Prospective cohort analysis. SETTING Administrative database of the US Veteran Affairs, 2002-6. Population 819 491 predominantly male participants (98%) aged 65 or more with cardiovascular disease. MAIN OUTCOME MEASURES Time to incident Alzheimer's disease or dementia in three cohorts (angiotensin receptor blockers, lisinopril, and other cardiovascular drugs, the "cardiovascular comparator") over a four year period (fiscal years 2003-6) using Cox proportional hazard models with adjustments for age, diabetes, stroke, and cardiovascular disease. Disease progression was the time to admission to a nursing home or death among participants with pre-existing Alzheimer's disease or dementia. RESULTS Hazard rates for incident dementia in the angiotensin receptor blocker group were 0.76 (95% confidence interval 0.69 to 0.84) compared with the cardiovascular comparator and 0.81 (0.73 to 0.90) compared with the lisinopril group. Compared with the cardiovascular comparator, angiotensin receptor blockers in patients with pre-existing Alzheimer's disease were associated with a significantly lower risk of admission to a nursing home (0.51, 0.36 to 0.72) and death (0.83, 0.71 to 0.97). Angiotensin receptor blockers exhibited a dose-response as well as additive effects in combination with angiotensin converting enzyme inhibitors. This combination compared with angiotensin converting enzyme inhibitors alone was associated with a reduced risk of incident dementia (0.54, 0.51 to 0.57) and admission to a nursing home (0.33, 0.22 to 0.49). Minor differences were shown in mean systolic and diastolic blood pressures between the groups. Similar results were observed for Alzheimer's disease. CONCLUSIONS Angiotensin receptor blockers are associated with a significant reduction in the incidence and progression of Alzheimer's disease and dementia compared with angiotensin converting enzyme inhibitors or other cardiovascular drugs in a predominantly male population.
Collapse
Affiliation(s)
- Nien-Chen Li
- Center for the Assessment of Pharmaceutical Practices and Pharmaceutical Assessment, Management and Policy Program, Department of Health Policy and Management, Boston University School of Public Health, Boston, MA 02118-2526, USA
| | | | | | | | | | | | | |
Collapse
|
237
|
Xu Z, Cacatian S, Yuan J, Simpson RD, Jia L, Zhao W, Tice CM, Flaherty PT, Guo J, Ishchenko A, Singh SB, Wu Z, McKeever BM, Scott BB, Bukhtiyarov Y, Berbaum J, Mason J, Panemangalore R, Cappiello MG, Bentley R, Doe CP, Harrison RK, McGeehan GM, Dillard LW, Baldwin JJ, Claremon DA. Optimization of orally bioavailable alkyl amine renin inhibitors. Bioorg Med Chem Lett 2010; 20:694-9. [DOI: 10.1016/j.bmcl.2009.11.066] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 11/11/2009] [Accepted: 11/16/2009] [Indexed: 11/27/2022]
|
238
|
Noh HJ, Kang YS, Lim DY. Effects of losartan on catecholamine release in the isolated rat adrenal gland. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2009; 13:327-35. [PMID: 19885018 DOI: 10.4196/kjpp.2009.13.4.327] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Revised: 08/03/2009] [Accepted: 08/17/2009] [Indexed: 11/15/2022]
Abstract
The aim of this study was to determine whether losartan, an angiotensin II (Ang II) type 1 (AT(1)) receptor could influence the CA release from the isolated perfused model of the rat adrenal medulla. Losartan (5~50 microM) perfused into an adrenal vein for 90 min produced dose- and time-dependent inhibition of the CA secretory responses evoked by ACh (5.32 mM), high K(+) (56 mM, a direct membrane depolarizer), DMPP (100 microM) and McN-A-343 (100 microM). Losartan failed to affect basal CA output. Furthermore, in adrenal glands loaded with losartan (15 microM) for 90 min, the CA secretory responses evoked by Bay-K-8644 (10 microM, an activator of L-type Ca(2+) channels), cyclopiazonic acid (10 microM, an inhibitor of cytoplasmic Ca(2+)-ATPase), veratridine (100 microM, an activator of Na(+) channels), and Ang II (100 nM) were markedly inhibited. However, at high concentrations (150~300 microM), losartan rather enhanced the CA secretion evoked by ACh. Collectively, these experimental results suggest that losartan at low concentrations inhibits the CA secretion evoked by cholinergic stimulation (both nicotininc and muscarinic receptors) as well as by membrane depolarization from the rat adrenal medulla, but at high concentration it rather inhibits ACh-evoked CA secretion. It seems that losartan has a dual action, acting as both agonist and antagonist to nicotinic receptors of the rat adrenal medulla, which might be dependent on the concentration. It is also thought that this inhibitory effect of losartan may be mediated by blocking the influx of both Na(+) and Ca(2+) into the rat adrenomedullary chromaffin cells as well as by inhibiting the Ca(2+) release from the cytoplasmic calcium store, which is thought to be relevant to the AT(1) receptor blockade, in addition to its enhancement of the CA release.
Collapse
Affiliation(s)
- Hae-Jeong Noh
- Department of Family Medicine, Eulji University Hospital, Daejeon 302-799
| | | | | |
Collapse
|
239
|
Wu J, Aluko RE, Muir AD. Production of angiotensin I-converting enzyme inhibitory peptides from defatted canola meal. BIORESOURCE TECHNOLOGY 2009; 100:5283-5287. [PMID: 19570676 DOI: 10.1016/j.biortech.2009.03.090] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2008] [Revised: 03/02/2009] [Accepted: 03/02/2009] [Indexed: 05/28/2023]
Abstract
To simplify the method of ACE-inhibitory peptide production, defatted canola meal was subjected to enzymatic proteolysis. Alcalase 2.4 L and protease M "Amano" were found to be the most efficient enzymes in releasing ACE-inhibitory peptides from canola proteins among 13 tested enzymes. The IC(50) values of canola protein hydrolysates ranged from 18.1 to 82.5 microg protein/mL. Differences in ACE-inhibitory activities of various protein hydrolysates reflected varied enzyme specificities. A positive correlation was determined between ACE-inhibitory activity and the degree of hydrolysis (r=0.5916, p<0.001). Ion-exchange chromatography of canola protein hydrolysate increased the protein content greater than 95% without loss of ACE-inhibitory activity. This fraction was resistant to the degradation of gastrointestinal enzyme and ACE during in vitro incubation and may be a useful ingredient in the formulation of hypotensive functional food products.
Collapse
Affiliation(s)
- Jianping Wu
- Agriculture and Agri-Food Canada, Saskatoon Research Station, 107 Science Place, Saskatoon, SK, Canada S7N OX2.
| | | | | |
Collapse
|
240
|
Multivalent ligand-receptor interactions elicit inverse agonist activity of AT(1) receptor blockers against stretch-induced AT(1) receptor activation. Hypertens Res 2009; 32:875-83. [PMID: 19662020 DOI: 10.1038/hr.2009.117] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Type 1 angiotensin II (AT(1)) receptor has a critical role in the development of load-induced cardiac hypertrophy. Recently, we showed that mechanical stretching of cells activates the AT(1) receptor without the involvement of angiotensin II (AngII) and that this AngII-independent activation is inhibited by the inverse agonistic activity of the AT(1) receptor blocker (ARB), candesartan. Although the inverse agonist activity of ARBs has been studied in terms of their action on constitutively active AT(1) receptors, the structure-function relationship of the inverse agonism they exert against stretch-induced AT(1) receptor activation has not been fully elucidated. Assays evaluating c-fos gene expression and phosphorylated extracellular signal-regulated protein kinases (ERKs) have shown that olmesartan has strong inverse agonist activities against the constitutively active AT(1) receptor and the stretch-induced activation of AT(1) receptor, respectively. Ternary drug-receptor interactions, which occur between the hydroxyl group of olmesartan and Tyr(113) and between the carboxyl group of olmesartan and Lys(199) and His(256), were essential for the potent inverse agonist action olmesartan exerts against stretch-induced ERK activation and the constitutive activity of the AT(1)-N111G mutant receptor. Furthermore, the inverse agonist activity olmesartan exerts against stretch-induced ERK activation requires an additional drug-receptor interaction involving the tetrazole group of olmesartan and Gln(257) of the AT(1) receptor. These results suggest that multivalent interactions between an inverse agonist and the AT(1) receptor are required to stabilize the receptor in an inactive conformation in response to the distinct processes that lead to an AngII-independent activation of the AT(1) receptor.
Collapse
|
241
|
Abstract
Proteases constitute one of the primary targets in drug discovery. In the present review, we focus on extracellular proteases (ECPs) because of their differential expression in many pathophysiological processes, including cancer, cardiovascular conditions, and inflammatory, pulmonary, and periodontal diseases. Many new ECP inhibitors are currently under clinical investigation and a significant increase in new therapies based on protease inhibition can be expected in the coming years. In addition to directly blocking the activity of a targeted protease, one can take advantage of differential expression in disease states to selectively deliver therapeutic or imaging agents. Recent studies in targeted drug development for the metalloproteases (matrix metalloproteinases, adamalysins, pappalysins, neprilysin, angiotensin-converting enzyme, metallocarboxypeptidases, and glutamate carboxypeptidase II), serine proteases (elastase, coagulation factors, tissue/urokinase plasminogen activator system, kallikreins, tryptase, dipeptidyl peptidase IV) and cysteine proteases (cathepsin B) are discussed herein.
Collapse
Affiliation(s)
- Mare Cudic
- Department of Chemistry & Biochemistry, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431 U.S.A
| | - Gregg B. Fields
- Department of Biochemistry, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229 U.S.A
| |
Collapse
|
242
|
Barbalic M, Schwartz GL, Chapman AB, Turner ST, Boerwinkle E. Kininogen gene (KNG) variation has a consistent effect on aldosterone response to antihypertensive drug therapy: the GERA study. Physiol Genomics 2009; 39:56-60. [PMID: 19584173 DOI: 10.1152/physiolgenomics.00061.2009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recent experimental and clinical studies suggested that apart from playing an essential role in blood pressure homeostasis, aldosterone is involved in the pathophysiology of cardiovascular and renal diseases by inducing structural changes in the heart, kidney, and vessel wall. The interindividual variation of aldosterone response to antihypertensive treatment is considerable, and is at least partially explained by genetic variation. In this study, we investigated aldosterone response to two antihypertensive drugs-a thiazide diuretic and an angiotensin receptor blocker (ARB). Genetic variations in 50 candidate genes were tested for association with aldosterone response in four independent samples: African American (AA) responders to a diuretic (n = 289), AA responders to an ARB (n = 252), European American (EA) responders to a diuretic (n = 295) and EA responders to an ARB (n = 300). Linear regression was used to test the association with inclusion of age, sex, and body mass index as covariates. The results indicated the existence of one or more variants in the kininogen gene (KNG) that influence interindividual variation in aldosterone response. The significant association was replicated in three of four studied groups. The single nucleotide polymorphism rs4686799 was associated in AA and EA responders to the diuretic (P = 0.04 and P = 0.07, respectively), and rs5030062 and rs698078 were significantly associated in EA responders to the diuretic (P = 0.05 and P = 0.01) and EA responders to the ARB (P = 0.04 and P = 0.02). Although the clinical implication of KNG gene variation to antihypertensive drug response is yet to be determined, this novel candidate locus provides important new insights into drug response physiology.
Collapse
Affiliation(s)
- Maja Barbalic
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
243
|
Tice CM, Xu Z, Yuan J, Simpson RD, Cacatian ST, Flaherty PT, Zhao W, Guo J, Ishchenko A, Singh SB, Wu Z, Scott BB, Bukhtiyarov Y, Berbaum J, Mason J, Panemangalore R, Cappiello MG, Müller D, Harrison RK, McGeehan GM, Dillard LW, Baldwin JJ, Claremon DA. Design and optimization of renin inhibitors: Orally bioavailable alkyl amines. Bioorg Med Chem Lett 2009; 19:3541-5. [DOI: 10.1016/j.bmcl.2009.04.140] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Revised: 04/27/2009] [Accepted: 04/30/2009] [Indexed: 11/25/2022]
|
244
|
Pereira RM, Santos RASD, Dias FLDC, Teixeira MM, Silva ACSE. Renin-angiotensin system in the pathogenesis of liver fibrosis. World J Gastroenterol 2009; 15:2579-2586. [PMID: 19496186 PMCID: PMC2691487 DOI: 10.3748/wjg.15.2579] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2008] [Revised: 05/05/2009] [Accepted: 05/12/2009] [Indexed: 02/06/2023] Open
Abstract
Hepatic fibrosis is considered a common response to many chronic hepatic injuries. It is a multifunctional process that involves several cell types, cytokines, chemokines and growth factors leading to a disruption of homeostatic mechanisms that maintain the liver ecosystem. In spite of many studies regarding the development of fibrosis, the understanding of the pathogenesis remains obscure. The hepatic tissue remodeling process is highly complex, resulting from the balance between collagen degradation and synthesis. Among the many mediators that take part in this process, the components of the Renin angiotensin system (RAS) have progressively assumed an important role. Angiotensin (Ang) II acts as a profibrotic mediator and Ang-(1-7), the newly recognized RAS component, appears to exert a counter-regulatory role in liver tissue. We briefly review the liver fibrosis process and current aspects of the RAS. This review also aims to discuss some experimental evidence regarding the participation of RAS mediators in the pathogenesis of liver fibrosis, focusing on the putative role of the ACE2-Ang-(1-7)-Mas receptor axis.
Collapse
|
245
|
Pinheiro SVB, Ferreira AJ, Kitten GT, da Silveira KD, da Silva DA, Santos SHS, Gava E, Castro CH, Magalhães JA, da Mota RK, Botelho-Santos GA, Bader M, Alenina N, Santos RAS, Simoes E Silva AC. Genetic deletion of the angiotensin-(1-7) receptor Mas leads to glomerular hyperfiltration and microalbuminuria. Kidney Int 2009; 75:1184-1193. [PMID: 19262461 DOI: 10.1038/ki.2009.61] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Angiotensin-(1-7), an active fragment of both angiotensins I and II, generally opposes the vascular and proliferative actions of angiotensin II. Here we evaluated effects of the angiotensin-(1-7) receptor Mas on renal physiology and morphology using Mas-knockout mice. Compared to the wild-type animals, Mas knockout mice had significant reductions in urine volume and fractional sodium excretion without any significant change in free-water clearance. A significantly higher inulin clearance and microalbuminuria concomitant with a reduced renal blood flow suggest that glomerular hyperfiltration occurs in the knockout mice. Histological analysis found reduced glomerular tuft diameter and increased expression of collagen IV and fibronectin in the both the mesangium and interstitium, along with increased collagen III in the interstitium. These fibrogenic changes and the renal dysfunction of the knockout mice were associated with an upregulation of angiotensin II AT1 receptor and transforming growth factor-beta mRNA. Our study suggests that Mas acts as a critical regulator of renal fibrogenesis by controlling effects transduced through angiotensin II AT1 receptors in the kidney.
Collapse
Affiliation(s)
- Sérgio V B Pinheiro
- Pediatric Nephrology Unit, Medicine Faculty, Department of Pediatrics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Anderson J Ferreira
- Department of Morphology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Gregory T Kitten
- Department of Morphology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Kátia D da Silveira
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Deivid A da Silva
- Pediatric Nephrology Unit, Medicine Faculty, Department of Pediatrics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Sérgio H S Santos
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Elisandra Gava
- Department of Morphology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Carlos H Castro
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Júnio A Magalhães
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Renata K da Mota
- Department of Morphology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Giancarla A Botelho-Santos
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | - Robson A S Santos
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ana Cristina Simoes E Silva
- Pediatric Nephrology Unit, Medicine Faculty, Department of Pediatrics, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
246
|
Akazawa H, Yasuda N, Komuro I. Mechanisms and functions of agonist-independent activation in the angiotensin II type 1 receptor. Mol Cell Endocrinol 2009; 302:140-7. [PMID: 19059460 DOI: 10.1016/j.mce.2008.11.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Revised: 11/06/2008] [Accepted: 11/06/2008] [Indexed: 11/23/2022]
Abstract
The angiotensin II (AngII) type 1 (AT(1)) receptor is a seven-transmembrane G protein-coupled receptor, and is involved in regulating the physiological and pathological process of the cardiovascular system. Systemically and locally generated AngII has agonistic action on AT(1) receptor, but recent studies have demonstrated that AT(1) receptor inherently shows spontaneous activity even in the absence of AngII. Furthermore, mechanical stress can activate AT(1) receptor by inducing conformational switch without the involvement of AngII, and induce cardiac hypertrophy in vivo. These agonist-independent activities of AT(1) receptor can be inhibited by inverse agonists, but not by neutral antagonists. Considerable attention has been directed to molecular mechanisms and clinical implications of agonist-independent AT(1) receptor activation, and inverse agonist activity emerges as an important pharmacological parameter for AT(1) receptor blockers that will improve efficacy and expand therapeutic potentials in cardiovascular medicine.
Collapse
Affiliation(s)
- Hiroshi Akazawa
- Division of Cardiovascular Pathophysiology, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan
| | | | | |
Collapse
|
247
|
Lyngsø C, Erikstrup N, Hansen JL. Functional interactions between 7TM receptors in the renin-angiotensin system--dimerization or crosstalk? Mol Cell Endocrinol 2009; 302:203-12. [PMID: 18930783 DOI: 10.1016/j.mce.2008.09.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2008] [Revised: 09/09/2008] [Accepted: 09/10/2008] [Indexed: 12/15/2022]
Abstract
The Renin-Angiotensin System (RAS) is important for the regulation of cardiovascular physiology, where it controls blood pressure, and salt- and water homeostasis. Dysregulation of RAS can lead to severe diseases including hypertension, diabetic nephropathy, and cardiac arrhythmia, and -failure. The importance of the RAS is clearly emphasised by the widespread use of drugs targeting this system in clinical practice. These include, renin inhibitors, angiotensin II receptor type I blockers, and inhibitors of the angiotensin converting enzyme. Some of the important effectors within the system are 7 transmembrane (7TM) receptors (or G-protein-coupled receptors) such as the angiotensin II Receptors type I and II (AT1R and AT2R) and the MAS-oncogene receptor. Several findings indicate that the 7TM receptors can form both homo- and heterodimers, or higher orders of oligomers. Furthermore, dimerization may be important for receptor function, and in the development of cardiovascular diseases. This is very significant, since "dimers" may provide pharmacologists with novel targets for improved drug therapy. However, we know that 7TM receptors can mediate signals as monomeric units, and so far it has been very difficult to establish if our observations reflect actual well-defined dimerization or merely reflect close proximity between the receptors and/or various types of functional interaction. In this review, we will present and critically discuss the current data on 7TM receptor dimerization with a clear focus on the RAS, and delineate future challenges within the field.
Collapse
Affiliation(s)
- Christina Lyngsø
- Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | | |
Collapse
|
248
|
Masson R, Nicklin SA, Craig MA, McBride M, Gilday K, Gregorevic P, Allen JM, Chamberlain JS, Smith G, Graham D, Dominiczak AF, Napoli C, Baker AH. Onset of experimental severe cardiac fibrosis is mediated by overexpression of Angiotensin-converting enzyme 2. Hypertension 2009; 53:694-700. [PMID: 19221212 DOI: 10.1161/hypertensionaha.108.122333] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Angiotensin-converting enzyme (ACE) 2 is a recently identified homologue of ACE. There is great interest in the therapeutic benefit for ACE2 overexpression in the heart. However, the role of ACE2 in the regulation of cardiac structure and function, as well as maintenance of systemic blood pressure, remains poorly understood. In cell culture, ACE2 overexpression led to markedly increased myocyte volume, assessed in primary rabbit myocytes. To assess ACE2 function in vivo, we used a recombinant adeno-associated virus 6 delivery system to provide 11-week overexpression of ACE2 in the myocardium of stroke-prone spontaneously hypertensive rats. ACE2, as well as the ACE inhibitor enalapril, significantly reduced systolic blood pressure. However, in the heart, ACE2 overexpression resulted in cardiac fibrosis, as assessed by histological analysis with concomitant deficits in ejection fraction and fractional shortening measured by echocardiography. Furthermore, global gene expression profiling demonstrated the activation of profibrotic pathways in the heart mediated by ACE2 gene delivery. This study demonstrates that sustained overexpression of ACE2 in the heart in vivo leads to the onset of severe fibrosis.
Collapse
Affiliation(s)
- Rachel Masson
- BHF GCRC, University of Glasgow, 126 University Place, Glasgow, G12 8TA United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
249
|
Sun X, Wiesner B, Lorenz D, Papsdorf G, Pankow K, Wang P, Dietrich N, Siems WE, Maul B. Interaction of angiotensin-converting enzyme (ACE) with membrane-bound carboxypeptidase M (CPM) - a new function of ACE. Biol Chem 2009; 389:1477-85. [PMID: 18844448 DOI: 10.1515/bc.2008.168] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Angiotensin-converting enzyme (ACE) demonstrates, besides its typical dipeptidyl-carboxypeptidase activity, several unusual functions. Here, we demonstrate with molecular, biochemical, and cellular techniques that the somatic wild-type murine ACE (mACE), stably transfected in Chinese Hamster Ovary (CHO) or Madin-Darby Canine Kidney (MDCK) cells, interacts with endogenous membranal co-localized carboxypeptidase M (CPM). CPM belongs to the group of glycosylphosphatidylinositol (GPI)-anchored proteins. Here we report that ACE, completely independent of its known dipeptidase activities, has GPI-targeted properties. Our results indicate that the spatial proximity between mACE and the endogenous CPM enables an ACE-evoked release of CPM. These results are discussed with respect to the recently proposed GPI-ase activity and function of sperm-bound ACE.
Collapse
Affiliation(s)
- Xiaoou Sun
- Leibniz-Institut für Molekulare Pharmakologie, D-13125 Berlin, Germany and Charité-Universitätsmedizin Berlin, D-10117 Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
250
|
Hansen JL, Hansen JT, Speerschneider T, Lyngsø C, Erikstrup N, Burstein ES, Weiner DM, Walther T, Makita N, Iiri T, Merten N, Kostenis E, Sheikh SP. Lack of Evidence for AT1R/B2R Heterodimerization in COS-7, HEK293, and NIH3T3 Cells. J Biol Chem 2009; 284:1831-9. [PMID: 19017652 DOI: 10.1074/jbc.m804607200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Jakob L Hansen
- Laboratory for Molecular Cardiology, The Danish National Research Foundation Centre for Cardiac Arrhythmia, The Heart Centre, Copenhagen University Hospital, Copenhagen, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|