1
|
Cozier G, Coulson LB, Eyermann CJ, Basarab GS, Schwager SL, Chibale K, Sturrock ED, Acharya KR. Design of Novel Mercapto-3-phenylpropanoyl Dipeptides as Dual Angiotensin-Converting Enzyme C-Domain-Selective/Neprilysin Inhibitors. J Med Chem 2025; 68:7720-7736. [PMID: 40168649 PMCID: PMC11997989 DOI: 10.1021/acs.jmedchem.5c00329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/03/2025]
Abstract
Dual angiotensin-converting enzyme (ACE) and neprilysin (NEP) inhibitors such as omapatrilat showed promise as potent treatments for hypertension but produced adverse effects due to their high affinity for both domains of ACE (nACE and cACE). This led to the search for compounds that retained NEP potency but selectively inhibit cACE, leaving nACE active to degrade other peptides such as bradykinin. Lisinopril-tryptophan (LisW) has previously been reported to have cACE selectivity. Three mercapto-3-phenylpropanoyl inhibitors were synthesized, combining features of omapatrilat and LisW to probe structural characteristics required for potent dual cACE/NEP inhibition. We report the synthesis of these inhibitors, enzyme inhibition data, and high-resolution crystal structures in complex with nACE and cACE. This provides valuable insight into factors driving potency and selectivity and shows that the mercapto-3-phenylpropanoyl backbone is significantly better for NEP potency than a P1 carboxylate. Future chemistry efforts could be directed at identifying alternative chemotypes for optimization of cACE/NEP inhibitors.
Collapse
Affiliation(s)
- Gyles
E. Cozier
- Department
of Life Sciences, University of Bath, Claverton Down, Bath BA2
7AY, United Kingdom
| | - Lauren B. Coulson
- Institute
of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, South Africa
- Department
of Integrative Biomedical Sciences, University
of Cape Town, Observatory 7925, South Africa
| | - Charles J. Eyermann
- Drug
Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Gregory S. Basarab
- Drug
Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Sylva L. Schwager
- Institute
of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, South Africa
- Department
of Integrative Biomedical Sciences, University
of Cape Town, Observatory 7925, South Africa
| | - Kelly Chibale
- Institute
of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, South Africa
- Drug
Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa
- Department
of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- South
African Medical Research Council Drug Discovery and Development Research
Unit, University of Cape Town, Rondebosch 7701, South Africa
| | - Edward D. Sturrock
- Institute
of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, South Africa
- Department
of Integrative Biomedical Sciences, University
of Cape Town, Observatory 7925, South Africa
| | - K. Ravi Acharya
- Department
of Life Sciences, University of Bath, Claverton Down, Bath BA2
7AY, United Kingdom
| |
Collapse
|
2
|
Acharya K, Gregory K, Sturrock E. Advances in the structural basis for angiotensin-1 converting enzyme (ACE) inhibitors. Biosci Rep 2024; 44:BSR20240130. [PMID: 39046229 PMCID: PMC11300679 DOI: 10.1042/bsr20240130] [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: 05/31/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 07/25/2024] Open
Abstract
Human somatic angiotensin-converting enzyme (ACE) is a key zinc metallopeptidase that plays a pivotal role in the renin-angiotensin-aldosterone system (RAAS) by regulating blood pressure and electrolyte balance. Inhibition of ACE is a cornerstone in the management of hypertension, cardiovascular diseases, and renal disorders. Recent advances in structural biology techniques have provided invaluable insights into the molecular mechanisms underlying ACE inhibition, facilitating the design and development of more effective therapeutic agents. This review focuses on the latest advancements in elucidating the structural basis for ACE inhibition. High-resolution crystallographic studies of minimally glycosylated individual domains of ACE have revealed intricate molecular details of the ACE catalytic N- and C-domains, and their detailed interactions with clinically relevant and newly designed domain-specific inhibitors. In addition, the recently elucidated structure of the glycosylated form of full-length ACE by cryo-electron microscopy (cryo-EM) has shed light on the mechanism of ACE dimerization and revealed continuous conformational changes which occur prior to ligand binding. In addition to these experimental techniques, computational approaches have also played a pivotal role in elucidating the structural basis for ACE inhibition. Molecular dynamics simulations and computational docking studies have provided atomic details of inhibitor binding kinetics and energetics, facilitating the rational design of novel ACE inhibitors with improved potency and selectivity. Furthermore, computational analysis of the motions observed by cryo-EM allowed the identification of allosteric binding sites on ACE. This affords new opportunities for the development of next-generation allosteric inhibitors with enhanced pharmacological properties. Overall, the insights highlighted in this review could enable the rational design of novel ACE inhibitors with improved efficacy and safety profiles, ultimately leading to better therapeutic outcomes for patients with hypertension and cardiovascular diseases.
Collapse
Affiliation(s)
- K. Ravi Acharya
- Department of Life Sciences, University of Bath, Claverton Down, Bath BA2 7AY, U.K
| | - Kyle S. Gregory
- Department of Life Sciences, University of Bath, Claverton Down, Bath BA2 7AY, U.K
| | - Edward D. Sturrock
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Cape Town, Republic of South Africa
| |
Collapse
|
3
|
Trentin-Sonoda M, Cheff V, Gutsol A, Hébert RL. Sex-dependent effects of Canagliflozin on kidney protection in mice with combined hypertension-type 1 diabetes. PLoS One 2023; 18:e0295284. [PMID: 38055691 DOI: 10.1371/journal.pone.0295284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/17/2023] [Indexed: 12/08/2023] Open
Abstract
Canagliflozin (CANA) is a sodium-glucose cotransporter 2 (SGLT2) inhibitor with blood glucose lowering effects. CANA also promotes kidney protection in patients with cardiovascular diseases and type 2 diabetes (T2D), as well as in normoglycemic patients with hypertension or heart failure. Clinical studies, although conduct in both sexes, do not report sex-dependent differences in T2DM treated with CANA. However, the impact of CANA in type 1 diabetes, as well in sex-dependent outcomes in such cohort needs further understanding. To analyze the effects of CANA in mice with combined hypertension and type 1 diabetes, diabetes was induced by STZ injection (5 days, 50mg/kg/day) in both male and female 8 weeks old genetic hypertensive mice (Lin), whereas the control (Lin) received 0.1M sodium citrate injections. 8 weeks after STZ. Mice were fed either regular or CANA-infused diet for 4 weeks. 8 weeks after STZ, hyperglycemia was present in both male and female mice. CANA reversed BG increase mice fed regular diet. Male LinSTZ mice had elevated water intake, urine output, urinary albumin to creatinine ratio (ACR), kidney lesion score, and creatinine clearance compared to the Lin control group. Kidney injury was improved in male LinSTZ + CANA group in male mice. Water intake and urine output were not statistically significantly different in female LinSTZ compared to female LinSTZ+ CANA. Moreover, CANA did not improve kidney injury in female mice, showing no effect in creatinine clearance, lesion score and fibrosis when compared to LinSTZ fed regular diet. Here we show that Canagliflozin might exert different kidney protection effects in male compared to female mice with hypertension and type 1 diabetes. Sex-dimorphisms were previously found in the pathophysiology of diabetes induced by STZ. Therefore, we highlight the importance of in-depth investigation on sex-dependent effects of CANA, taking in consideration the unique characteristics of disease progression for each sex.
Collapse
Affiliation(s)
- Mayra Trentin-Sonoda
- Kidney Research Centre, Division of Nephrology, Department of Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Véronique Cheff
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Alex Gutsol
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Richard L Hébert
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
4
|
Alves-Lopes R, Lacchini S, Neves KB, Harvey A, Montezano AC, Touyz RM. Vasoprotective effects of NOX4 are mediated via polymerase and transient receptor potential melastatin 2 cation channels in endothelial cells. J Hypertens 2023; 41:1389-1400. [PMID: 37272080 PMCID: PMC10399938 DOI: 10.1097/hjh.0000000000003478] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND NOX4 activation has been implicated to have vasoprotective and blood pressure (BP)-lowering effects. Molecular mechanisms underlying this are unclear, but NOX4-induced regulation of the redox-sensitive Ca 2+ channel TRPM2 and effects on endothelial nitric oxide synthase (eNOS)-nitric oxide signalling may be important. METHOD Wild-type and LinA3, renin-expressing hypertensive mice, were crossed with NOX4 knockout mice. Vascular function was measured by myography. Generation of superoxide (O 2- ) and hydrogen peroxide (H 2 O 2 ) were assessed by lucigenin and amplex red, respectively, and Ca 2+ influx by Cal-520 fluorescence in rat aortic endothelial cells (RAEC). RESULTS BP was increased in NOX4KO, LinA3 and LinA3/NOX4KO mice. This was associated with endothelial dysfunction and vascular remodelling, with exaggerated effects in NOX4KO groups. The TRPM2 activator, ADPR, improved vascular relaxation in LinA3/NOX4KO mice, an effect recapitulated by H 2 O 2 . Inhibition of PARP and TRPM2 with olaparib and 2-APB, respectively, recapitulated endothelial dysfunction in NOX4KO. In endothelial cells, Ang II increased H 2 O 2 generation and Ca 2+ influx, effects reduced by TRPM2 siRNA, TRPM2 inhibitors (8-br-cADPR, 2-APB), olaparib and GKT137831 (NOX4 inhibitor). Ang II-induced eNOS activation was blocked by NOX4 and TRPM2 siRNA, GKT137831, PEG-catalase and 8-br-cADPR. CONCLUSION Our findings indicate that NOX4-induced H 2 O 2 production activates PARP/TRPM2, Ca 2+ influx, eNOS activation and nitric oxide release in endothelial cells. NOX4 deficiency impairs Ca 2+ homeostasis leading to endothelial dysfunction, an effect exacerbated in hypertension. We define a novel pathway linking endothelial NOX4/H 2 O 2 to eNOS/nitric oxide through PARP/TRPM2/Ca 2+ . This vasoprotective pathway is perturbed when NOX4 is downregulated and may have significance in conditions associated with endothelial dysfunction, including hypertension.
Collapse
Affiliation(s)
- Rheure Alves-Lopes
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Silvia Lacchini
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Karla B. Neves
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Adam Harvey
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Augusto C. Montezano
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Rhian M. Touyz
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| |
Collapse
|
5
|
Gutsol AA, Hale TM, Thibodeau JF, Holterman CE, Nasrallah R, Correa JWN, Touyz RM, Kennedy CRJ, Burger D, Hébert RL, Burns KD. Comparative Analysis of Hypertensive Tubulopathy in Animal Models of Hypertension and Its Relevance to Human Pathology. Toxicol Pathol 2023; 51:160-175. [PMID: 37632371 DOI: 10.1177/01926233231191128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2023]
Abstract
Assessment of hypertensive tubulopathy for more than fifty animal models of hypertension in experimental pathology employs criteria that do not correspond to lesional descriptors for tubular lesions in clinical pathology. We provide a critical appraisal of experimental hypertension with the same approach used to estimate hypertensive renal tubulopathy in humans. Four models with different pathogenesis of hypertension were analyzed-chronic angiotensin (Ang) II-infused and renin-overexpressing (TTRhRen) mice, spontaneously hypertensive (SHR), and Goldblatt two-kidney one-clip (2K1C) rats. Mouse models, SHR, and the nonclipped kidney in 2K1C rats had no regular signs of hypertensive tubulopathy. Histopathology in animals was mild and limited to variations in the volume density of tubular lumen and epithelium, interstitial space, and interstitial collagen. Affected kidneys in animals demonstrated lesion values that are significantly different compared with healthy controls but correspond to mild damage if compared with hypertensive humans. The most substantial human-like hypertensive tubulopathy was detected in the clipped kidney of 2K1C rats. For the first time, our study demonstrated the regular presence of chronic progressive nephropathy (CPN) in relatively young mice and rats with induced hypertension. Because CPN may confound the assessment of rodent models of hypertension, proliferative markers should be used to verify nonhypertensive tubulopathy.
Collapse
Affiliation(s)
- Alex A Gutsol
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Taben M Hale
- The University of Arizona, Phoenix, Arizona, USA
| | | | | | | | | | | | - Chris R J Kennedy
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- University of Ottawa, Ottawa, Ontario, Canada
| | - Dylan Burger
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- University of Ottawa, Ottawa, Ontario, Canada
| | - Richard L Hébert
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- University of Ottawa, Ottawa, Ontario, Canada
| | - Kevin D Burns
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
6
|
Abolbaghaei A, Turner M, Thibodeau JF, Holterman CE, Kennedy CRJ, Burger D. The Proteome of Circulating Large Extracellular Vesicles in Diabetes and Hypertension. Int J Mol Sci 2023; 24:ijms24054930. [PMID: 36902363 PMCID: PMC10003702 DOI: 10.3390/ijms24054930] [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: 11/23/2022] [Revised: 02/14/2023] [Accepted: 02/23/2023] [Indexed: 03/08/2023] Open
Abstract
Hypertension and diabetes induce vascular injury through processes that are not fully understood. Changes in extracellular vesicle (EV) composition could provide novel insights. Here, we examined the protein composition of circulating EVs from hypertensive, diabetic and healthy mice. EVs were isolated from transgenic mice overexpressing human renin in the liver (TtRhRen, hypertensive), OVE26 type 1 diabetic mice and wild-type (WT) mice. Protein content was analyzed using liquid chromatography-mass spectrometry. We identified 544 independent proteins, of which 408 were found in all groups, 34 were exclusive to WT, 16 were exclusive to OVE26 and 5 were exclusive to TTRhRen mice. Amongst the differentially expressed proteins, haptoglobin (HPT) was upregulated and ankyrin-1 (ANK1) was downregulated in OVE26 and TtRhRen mice compared with WT controls. Conversely, TSP4 and Co3A1 were upregulated and SAA4 was downregulated exclusively in diabetic mice; and PPN was upregulated and SPTB1 and SPTA1 were downregulated in hypertensive mice, compared to WT mice. Ingenuity pathway analysis identified enrichment in proteins associated with SNARE signaling, the complement system and NAD homeostasis in EVs from diabetic mice. Conversely, in EVs from hypertensive mice, there was enrichment in semaphroin and Rho signaling. Further analysis of these changes may improve understanding of vascular injury in hypertension and diabetes.
Collapse
Affiliation(s)
- Akram Abolbaghaei
- Chronic Disease Program, Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, ON K1H 8M5, Canada
| | - Maddison Turner
- Chronic Disease Program, Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, ON K1H 8M5, Canada
| | - Jean-François Thibodeau
- Chronic Disease Program, Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, ON K1H 8M5, Canada
| | - Chet E. Holterman
- Chronic Disease Program, Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, ON K1H 8M5, Canada
| | - Christopher R. J. Kennedy
- Chronic Disease Program, Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, ON K1H 8M5, Canada
- Departments of Medicine and Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Dylan Burger
- Chronic Disease Program, Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, ON K1H 8M5, Canada
- Departments of Medicine and Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- School of Pharmaceutical Sciences, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Correspondence: ; Tel.: +1-613-562-5800 (ext. 8241)
| |
Collapse
|
7
|
High fat diet is protective against kidney injury in hypertensive-diabetic mice, but leads to liver injury. PLoS One 2023; 18:e0281123. [PMID: 36730247 PMCID: PMC9894391 DOI: 10.1371/journal.pone.0281123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/13/2023] [Indexed: 02/03/2023] Open
Abstract
Chronic kidney disease (CKD) is a worldwide health burden with increases risk of end-stage renal function if left untreated. CKD induced in the context of metabolic syndrome (MS) increases risks of hypertension, hyperglycemia, excess body fat and dyslipidemia. To test if combining a high-fat diet (HFD) regimen onto the hypertensive/ diabetic phenotype would mimic features of MS induced-CKD in mice, hyperglycemia was induced in genetically hypertensive mice (Lin), followed by HFD regimen. For that, 8-week-old male were subjected to streptozotocin (STZ) intraperitoneal (i.p.) injections (50 mg/kg, 5 days consecutive). LinSTZ were fed a 60% kCal HFD for 8 weeks. Lin mice treated with STZ developed polydipsia, became hypertensive and hyperglycemic. HFD induced weight gain, protected against glomerular hypertrophy, scarring, and albuminuria at endpoint compared to regular diet fed LinSTZ. On the other hand, HFD induced steatosis, liver fibrosis, inflammation, and increase in AST/ALT ratio, characteristics of non-alcoholic liver disease. Taken together, our results show that LinSTZ mice fed a HFD did not lead to a more robust model of MS-induced CKD, protected against kidney injury, but inducing liver damage. More studies are necessary to understand the kidney protective mechanisms of HFD when superimposed with hypertension and type 1 diabetes.
Collapse
|
8
|
Gutsol AA, Blanco P, Hale TM, Thibodeau JF, Holterman CE, Nasrallah R, Correa JWN, Afanasiev SA, Touyz RM, Kennedy CRJ, Burger D, Hébert RL, Burns KD. Comparative analysis of hypertensive nephrosclerosis in animal models of hypertension and its relevance to human pathology. Glomerulopathy. PLoS One 2022; 17:e0264136. [PMID: 35176122 PMCID: PMC8853553 DOI: 10.1371/journal.pone.0264136] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 02/03/2022] [Indexed: 01/09/2023] Open
Abstract
Current research on hypertension utilizes more than fifty animal models that rely mainly on stable increases in systolic blood pressure. In experimental hypertension, grading or scoring of glomerulopathy in the majority of studies is based on a wide range of opinion-based histological changes that do not necessarily comply with lesional descriptors for glomerular injury that are well-established in clinical pathology. Here, we provide a critical appraisal of experimental hypertensive glomerulopathy with the same approach used to assess hypertensive glomerulopathy in humans. Four hypertensive models with varying pathogenesis were analyzed–chronic angiotensin II infused mice, mice expressing active human renin in the liver (TTRhRen), spontaneously hypertensive rats (SHR), and Goldblatt two-kidney one-clip rats (2K1C). Analysis of glomerulopathy utilized the same criteria applied in humans–hyalinosis, focal segmental glomerulosclerosis (FSGS), ischemic, hypertrophic and solidified glomeruli, or global glomerulosclerosis (GGS). Data from animal models were compared to human reference values. Kidneys in TTRhRen mice, SHR and the nonclipped kidneys in 2K1C rats had no sign of hyalinosis, FSGS or GGS. Glomerulopathy in these groups was limited to variations in mesangial and capillary compartment volumes, with mild increases in collagen deposition. Histopathology in angiotensin II infused mice corresponded to mesangioproliferative glomerulonephritis, but not hypertensive glomerulosclerosis. The number of nephrons was significantly reduced in TTRhRen mice and SHR, but did not correlate with severity of glomerulopathy. The most substantial human-like glomerulosclerotic lesions, including FSGS, ischemic obsolescent glomeruli and GGS, were found in the clipped kidneys of 2K1C rats. The comparison of affected kidneys to healthy control in animals produces lesion values that are numerically impressive but correspond to mild damage if compared to humans. Animal studies should be standardized by employing the criteria and classifications established in human pathology to make experimental and human data fully comparable for comprehensive analysis and model improvements.
Collapse
Affiliation(s)
- Alex A. Gutsol
- Ottawa Hospital Research Institute & Kidney Research Centre, University of Ottawa, Ottawa, ON, Canada
- * E-mail:
| | - Paula Blanco
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Taben M. Hale
- Basic Medical Sciences Faculty, University of Arizona, Tucson, AZ, United States of America
| | - Jean-Francois Thibodeau
- Ottawa Hospital Research Institute & Kidney Research Centre, University of Ottawa, Ottawa, ON, Canada
| | - Chet E. Holterman
- Ottawa Hospital Research Institute & Kidney Research Centre, University of Ottawa, Ottawa, ON, Canada
| | - Rania Nasrallah
- Ottawa Hospital Research Institute & Kidney Research Centre, University of Ottawa, Ottawa, ON, Canada
| | - Jose W. N. Correa
- Department of Physiological Sciences, Biological Sciences Institute, Federal University of Amazonas, Manaus, Brazil
| | | | - Rhian M. Touyz
- Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Chris R. J. Kennedy
- Ottawa Hospital Research Institute & Kidney Research Centre, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Dylan Burger
- Ottawa Hospital Research Institute & Kidney Research Centre, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Richard L. Hébert
- Ottawa Hospital Research Institute & Kidney Research Centre, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Kevin D. Burns
- Ottawa Hospital Research Institute & Kidney Research Centre, University of Ottawa, Ottawa, ON, Canada
- Division of Nephrology, Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
9
|
Arendse LB, Cozier GE, Eyermann CJ, Basarab GS, Schwager SL, Chibale K, Acharya KR, Sturrock ED. Probing the Requirements for Dual Angiotensin-Converting Enzyme C-Domain Selective/Neprilysin Inhibition. J Med Chem 2022; 65:3371-3387. [PMID: 35113565 DOI: 10.1021/acs.jmedchem.1c01924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Selective inhibition of the angiotensin-converting enzyme C-domain (cACE) and neprilysin (NEP), leaving the ACE N-domain (nACE) free to degrade bradykinin and other peptides, has the potential to provide the potent antihypertensive and cardioprotective benefits observed for nonselective dual ACE/NEP inhibitors, such as omapatrilat, without the increased risk of adverse effects. We have synthesized three 1-carboxy-3-phenylpropyl dipeptide inhibitors with nanomolar potency based on the previously reported C-domain selective ACE inhibitor lisinopril-tryptophan (LisW) to probe the structural requirements for potent dual cACE/NEP inhibition. Here we report the synthesis, enzyme kinetic data, and high-resolution crystal structures of these inhibitors bound to nACE and cACE, providing valuable insight into the factors driving potency and selectivity. Overall, these results highlight the importance of the interplay between the S1' and S2' subsites for ACE domain selectivity, providing guidance for future chemistry efforts toward the development of dual cACE/NEP inhibitors.
Collapse
Affiliation(s)
- Lauren B Arendse
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town 7925, South Africa.,Department of Integrative Biomedical Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa
| | - Gyles E Cozier
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, U.K
| | - Charles J Eyermann
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch, Cape Town 7701, South Africa
| | - Gregory S Basarab
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch, Cape Town 7701, South Africa
| | - Sylva L Schwager
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town 7925, South Africa.,Department of Integrative Biomedical Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa
| | - Kelly Chibale
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town 7925, South Africa.,Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch, Cape Town 7701, South Africa.,Department of Chemistry, University of Cape Town, Rondebosch, Cape Town 7701, South Africa.,South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch, Cape Town 7701, South Africa
| | - K Ravi Acharya
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, U.K
| | - Edward D Sturrock
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town 7925, South Africa.,Department of Integrative Biomedical Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa
| |
Collapse
|
10
|
Goh NY, Mohamad Razif MF, Yap YHY, Ng CL, Fung SY. In silico analysis and characterization of medicinal mushroom cystathionine beta-synthase as an angiotensin converting enzyme (ACE) inhibitory protein. Comput Biol Chem 2021; 96:107620. [PMID: 34971900 DOI: 10.1016/j.compbiolchem.2021.107620] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 12/17/2022]
Abstract
Angiotensin-converting enzyme (ACE) regulates blood pressure and has been implicated in several conditions including lung injury, fibrosis and Alzheimer's disease. Medicinal mushroom Ganordema lucidum (Reishi) cystathionine beta-synthase (GlCBS) was previously reported to possess ACE inhibitory activities. However, the inhibitory mechanism of CBS protein remains unreported. Therefore, this study integrates in silico sequencing, structural and functional based-analysis, protein modelling, molecular docking and binding affinity calculation to elucidate the inhibitory mechanism of GlCBS and Lignosus rhinocerus (Tiger milk mushroom) CBS protein (LrCBS) towards ACE. In silico analysis indicates that CBSs from both mushrooms share high similarities in terms of physical properties, structural properties and domain distribution. Protein-protein docking analysis revealed that both GlCBS and LrCBS potentially modulate the C-terminal domain of ACE (C-ACE) activity via regulation of chloride activation and/or prevention of substrate entry. GICBS and LrCBS were also shown to interact with ACE at the same region that presumably inhibits the function of ACE.
Collapse
Affiliation(s)
- Neng-Yao Goh
- Medicinal Mushroom Research Group (MMRG), Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Muhammad Fazril Mohamad Razif
- Medicinal Mushroom Research Group (MMRG), Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yeannie Hui-Yeng Yap
- Department of Oral Biology and Biomedical Sciences, MAHSA University, Selangor, Malaysia
| | - Chyan Leong Ng
- Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia, 43600 UKM Bangi, Selangor, Malaysia.
| | - Shin-Yee Fung
- Medicinal Mushroom Research Group (MMRG), Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
11
|
Alves-Lopes R, Montezano AC, Neves KB, Harvey A, Rios FJ, Skiba DS, Arendse LB, Guzik TJ, Graham D, Poglitsch M, Sturrock E, Touyz RM. Selective Inhibition of the C-Domain of ACE (Angiotensin-Converting Enzyme) Combined With Inhibition of NEP (Neprilysin): A Potential New Therapy for Hypertension. Hypertension 2021; 78:604-616. [PMID: 34304582 PMCID: PMC8357049 DOI: 10.1161/hypertensionaha.121.17041] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 06/25/2021] [Indexed: 12/11/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Rhéure Alves-Lopes
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | - Augusto C. Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | - Karla B. Neves
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | - Adam Harvey
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | - Francisco J. Rios
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | - Dominik S. Skiba
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | - Lauren B. Arendse
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Biochemistry, University of Cape Town, South Africa (L.B.A., E.S.)
| | - Tomasz J. Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | - Delyth Graham
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | | | - Edward Sturrock
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Biochemistry, University of Cape Town, South Africa (L.B.A., E.S.)
| | - Rhian M. Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| |
Collapse
|
12
|
Sharma U, Cozier GE, Sturrock ED, Acharya KR. Molecular Basis for Omapatrilat and Sampatrilat Binding to Neprilysin-Implications for Dual Inhibitor Design with Angiotensin-Converting Enzyme. J Med Chem 2020; 63:5488-5500. [PMID: 32337993 PMCID: PMC7304895 DOI: 10.1021/acs.jmedchem.0c00441] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
![]()
Neprilysin
(NEP) and angiotensin-converting enzyme (ACE) are two
key zinc-dependent metallopeptidases in the natriuretic peptide and
kinin systems and renin–angiotensin–aldosterone system,
respectively. They play an important role in blood pressure regulation
and reducing the risk of heart failure. Vasopeptidase inhibitors omapatrilat
and sampatrilat possess dual activity against these enzymes by blocking
the ACE-dependent conversion of angiotensin I to the potent vasoconstrictor
angiotensin II while simultaneously halting the NEP-dependent degradation
of vasodilator atrial natriuretic peptide. Here, we report crystal
structures of omapatrilat, sampatrilat, and sampatrilat-ASP (a sampatrilat
analogue) in complex with NEP at 1.75, 2.65, and 2.6 Å, respectively.
A detailed analysis of these structures and the corresponding structures
of ACE with these inhibitors has provided the molecular basis of dual
inhibitor recognition involving the catalytic site in both enzymes.
This new information will be very useful in the design of safer and
more selective vasopeptidase inhibitors of NEP and ACE for effective
treatment in hypertension and heart failure.
Collapse
Affiliation(s)
- Urvashi Sharma
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, United Kingdom
| | - Gyles E Cozier
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, United Kingdom
| | - Edward D Sturrock
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, 7935 Cape Town, Republic of South Africa
| | - K Ravi Acharya
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, United Kingdom
| |
Collapse
|
13
|
Alves-Lopes R, Neves KB, Anagnostopoulou A, Rios FJ, Lacchini S, Montezano AC, Touyz RM. Crosstalk Between Vascular Redox and Calcium Signaling in Hypertension Involves TRPM2 (Transient Receptor Potential Melastatin 2) Cation Channel. Hypertension 2019; 75:139-149. [PMID: 31735084 DOI: 10.1161/hypertensionaha.119.13861] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Increased generation of reactive oxygen species (ROS) and altered Ca2+ handling cause vascular damage in hypertension. Mechanisms linking these systems are unclear, but TRPM2 (transient receptor potential melastatin 2) could be important because TRPM2 is a ROS sensor and a regulator of Ca2+ and Na+ transport. We hypothesized that TRPM2 is a point of cross-talk between redox and Ca2+ signaling in vascular smooth muscle cells (VSMC) and that in hypertension ROS mediated-TRPM2 activation increases [Ca2+]i through processes involving NCX (Na+/Ca2+ exchanger). VSMCs from hypertensive and normotensive individuals and isolated arteries from wild type and hypertensive mice (LinA3) were studied. Generation of superoxide anion and hydrogen peroxide (H2O2) was increased in hypertensive VSMCs, effects associated with activation of redox-sensitive PARP1 (poly [ADP-ribose] polymerase 1), a TRPM2 regulator. Ang II (angiotensin II) increased Ca2+ and Na+ influx with exaggerated responses in hypertension. These effects were attenuated by catalase-polyethylene glycol -catalase and TRPM2 inhibitors (2-APB, 8-Br-cADPR olaparib). TRPM2 siRNA decreased Ca2+ in hypertensive VSMCs. NCX inhibitors (Benzamil, KB-R7943, YM244769) normalized Ca2+ hyper-responsiveness and MLC20 phosphorylation in hypertensive VSMCs. In arteries from LinA3 mice, exaggerated agonist (U46619, Ang II, phenylephrine)-induced vasoconstriction was decreased by TRPM2 and NCX inhibitors. In conclusion, activation of ROS-dependent PARP1-regulated TRPM2 contributes to vascular Ca2+ and Na+ influx in part through NCX. We identify a novel pathway linking ROS to Ca2+ signaling through TRPM2/NCX in human VSMCs and suggest that oxidative stress-induced upregulation of this pathway may be a new player in hypertension-associated vascular dysfunction.
Collapse
Affiliation(s)
- Rhéure Alves-Lopes
- From the Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., K.B.N., A.A., F.J.R., A.C.M., R.M.T.)
| | - Karla B Neves
- From the Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., K.B.N., A.A., F.J.R., A.C.M., R.M.T.)
| | - Aikaterini Anagnostopoulou
- From the Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., K.B.N., A.A., F.J.R., A.C.M., R.M.T.)
| | - Francisco J Rios
- From the Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., K.B.N., A.A., F.J.R., A.C.M., R.M.T.)
| | - Silvia Lacchini
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo Medical School, Brazil (S.L.)
| | - Augusto C Montezano
- From the Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., K.B.N., A.A., F.J.R., A.C.M., R.M.T.)
| | - Rhian M Touyz
- From the Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., K.B.N., A.A., F.J.R., A.C.M., R.M.T.)
| |
Collapse
|
14
|
Arendse LB, Danser AHJ, Poglitsch M, Touyz RM, Burnett JC, Llorens-Cortes C, Ehlers MR, Sturrock ED. Novel Therapeutic Approaches Targeting the Renin-Angiotensin System and Associated Peptides in Hypertension and Heart Failure. Pharmacol Rev 2019; 71:539-570. [PMID: 31537750 PMCID: PMC6782023 DOI: 10.1124/pr.118.017129] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Despite the success of renin-angiotensin system (RAS) blockade by angiotensin-converting enzyme (ACE) inhibitors and angiotensin II type 1 receptor (AT1R) blockers, current therapies for hypertension and related cardiovascular diseases are still inadequate. Identification of additional components of the RAS and associated vasoactive pathways, as well as new structural and functional insights into established targets, have led to novel therapeutic approaches with the potential to provide improved cardiovascular protection and better blood pressure control and/or reduced adverse side effects. The simultaneous modulation of several neurohumoral mediators in key interconnected blood pressure-regulating pathways has been an attractive approach to improve treatment efficacy, and several novel approaches involve combination therapy or dual-acting agents. In addition, increased understanding of the complexity of the RAS has led to novel approaches aimed at upregulating the ACE2/angiotensin-(1-7)/Mas axis to counter-regulate the harmful effects of the ACE/angiotensin II/angiotensin III/AT1R axis. These advances have opened new avenues for the development of novel drugs targeting the RAS to better treat hypertension and heart failure. Here we focus on new therapies in preclinical and early clinical stages of development, including novel small molecule inhibitors and receptor agonists/antagonists, less conventional strategies such as gene therapy to suppress angiotensinogen at the RNA level, recombinant ACE2 protein, and novel bispecific designer peptides.
Collapse
Affiliation(s)
- Lauren B Arendse
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa (L.B.A., E.D.S.); Division of Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (A.H.J.D.); Attoquant Diagnostics, Vienna, Austria (M.P.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.M.T.); Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota (J.C.B.); Institut National de la Santé et de la Recherche Médicale, Paris, France (C.L.-C.); and Clinical Trials Group, Immune Tolerance Network, San Francisco, California (M.R.E.)
| | - A H Jan Danser
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa (L.B.A., E.D.S.); Division of Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (A.H.J.D.); Attoquant Diagnostics, Vienna, Austria (M.P.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.M.T.); Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota (J.C.B.); Institut National de la Santé et de la Recherche Médicale, Paris, France (C.L.-C.); and Clinical Trials Group, Immune Tolerance Network, San Francisco, California (M.R.E.)
| | - Marko Poglitsch
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa (L.B.A., E.D.S.); Division of Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (A.H.J.D.); Attoquant Diagnostics, Vienna, Austria (M.P.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.M.T.); Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota (J.C.B.); Institut National de la Santé et de la Recherche Médicale, Paris, France (C.L.-C.); and Clinical Trials Group, Immune Tolerance Network, San Francisco, California (M.R.E.)
| | - Rhian M Touyz
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa (L.B.A., E.D.S.); Division of Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (A.H.J.D.); Attoquant Diagnostics, Vienna, Austria (M.P.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.M.T.); Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota (J.C.B.); Institut National de la Santé et de la Recherche Médicale, Paris, France (C.L.-C.); and Clinical Trials Group, Immune Tolerance Network, San Francisco, California (M.R.E.)
| | - John C Burnett
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa (L.B.A., E.D.S.); Division of Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (A.H.J.D.); Attoquant Diagnostics, Vienna, Austria (M.P.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.M.T.); Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota (J.C.B.); Institut National de la Santé et de la Recherche Médicale, Paris, France (C.L.-C.); and Clinical Trials Group, Immune Tolerance Network, San Francisco, California (M.R.E.)
| | - Catherine Llorens-Cortes
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa (L.B.A., E.D.S.); Division of Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (A.H.J.D.); Attoquant Diagnostics, Vienna, Austria (M.P.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.M.T.); Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota (J.C.B.); Institut National de la Santé et de la Recherche Médicale, Paris, France (C.L.-C.); and Clinical Trials Group, Immune Tolerance Network, San Francisco, California (M.R.E.)
| | - Mario R Ehlers
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa (L.B.A., E.D.S.); Division of Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (A.H.J.D.); Attoquant Diagnostics, Vienna, Austria (M.P.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.M.T.); Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota (J.C.B.); Institut National de la Santé et de la Recherche Médicale, Paris, France (C.L.-C.); and Clinical Trials Group, Immune Tolerance Network, San Francisco, California (M.R.E.)
| | - Edward D Sturrock
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa (L.B.A., E.D.S.); Division of Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (A.H.J.D.); Attoquant Diagnostics, Vienna, Austria (M.P.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.M.T.); Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota (J.C.B.); Institut National de la Santé et de la Recherche Médicale, Paris, France (C.L.-C.); and Clinical Trials Group, Immune Tolerance Network, San Francisco, California (M.R.E.)
| |
Collapse
|
15
|
Structural basis for the C-domain-selective angiotensin-converting enzyme inhibition by bradykinin-potentiating peptide b (BPPb). Biochem J 2019; 476:1553-1570. [DOI: 10.1042/bcj20190290] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 04/15/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
Abstract
Abstract
Angiotensin-converting enzyme (ACE) is a zinc metalloprotease best known for its role in blood pressure regulation. ACE consists of two homologous catalytic domains, the N- and C-domain, that display distinct but overlapping catalytic functions in vivo owing to subtle differences in substrate specificity. While current generation ACE inhibitors target both ACE domains, domain-selective ACE inhibitors may be clinically advantageous, either reducing side effects or having utility in new indications. Here, we used site-directed mutagenesis, an ACE chimera and X-ray crystallography to unveil the molecular basis for C-domain-selective ACE inhibition by the bradykinin-potentiating peptide b (BPPb), naturally present in Brazilian pit viper venom. We present the BPPb N-domain structure in comparison with the previously reported BPPb C-domain structure and highlight key differences in peptide interactions with the S4 to S9 subsites. This suggests the involvement of these subsites in conferring C-domain-selective BPPb binding, in agreement with the mutagenesis results where unique residues governing differences in active site exposure, lid structure and dynamics between the two domains were the major drivers for C-domain-selective BPPb binding. Mere disruption of BPPb interactions with unique S2 and S4 subsite residues, which synergistically assist in BPPb binding, was insufficient to abolish C-domain selectivity. The combination of unique S9–S4 and S2′ subsite C-domain residues was required for the favourable entry, orientation and thus, selective binding of the peptide. This emphasizes the need to consider factors other than direct protein–inhibitor interactions to guide the design of domain-selective ACE inhibitors, especially in the case of larger peptides.
Collapse
|
16
|
AL Mulhim NS, Kehoe PG, Miners JS. Divergence in the activity of the N- and C- catalytic domains of ACE1 - implications for the role of the renin-angiotensin system in Alzheimer's disease. Acta Neuropathol Commun 2019; 7:57. [PMID: 31018864 PMCID: PMC6480511 DOI: 10.1186/s40478-019-0718-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 04/10/2019] [Indexed: 01/01/2023] Open
|
17
|
Thibodeau JF, Holterman CE, He Y, Carter A, Cron GO, Boisvert NC, Abd-Elrahman KS, Hsu KJ, Ferguson SSG, Kennedy CRJ. Vascular Smooth Muscle-Specific EP4 Receptor Deletion in Mice Exacerbates Angiotensin II-Induced Renal Injury. Antioxid Redox Signal 2016; 25:642-656. [PMID: 27245461 DOI: 10.1089/ars.2015.6592] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
AIMS Cyclooxygenase inhibition by non-steroidal anti-inflammatory drugs is contraindicated in hypertension, as it may reduce glomerular filtration rate (GFR) and renal blood flow. However, the identity of the specific eicosanoid and receptor underlying these effects is not known. We hypothesized that vascular smooth muscle prostaglandin E2 (PGE2) E-prostanoid 4 (EP4) receptor deletion predisposes to renal injury via unchecked vasoconstrictive actions of angiotensin II (AngII) in a hypertension model. Mice with inducible vascular smooth muscle cell (VSMC)-specific EP4 receptor deletion were generated and subjected to AngII-induced hypertension. RESULTS EP4 deletion was verified by PCR of aorta and renal vessels, as well as functionally by loss of PGE2-mediated mesenteric artery relaxation. Both AngII-treated groups became similarly hypertensive, whereas albuminuria, foot process effacement, and renal hypertrophy were exacerbated in AngII-treated EP4VSMC-/- but not in EP4VSMC+/+ mice and were associated with glomerular scarring, tubulointerstitial injury, and reduced GFR. AngII-treated EP4VSMC-/- mice exhibited capillary damage and reduced renal perfusion as measured by fluorescent bead microangiography and magnetic resonance imaging, respectively. NADPH oxidase 2 (Nox2) expression was significantly elevated in AngII-treated EP4-/- mice. EP4-receptor silencing in primary VSMCs abolished PGE2 inhibition of AngII-induced Nox2 mRNA and superoxide production. INNOVATION These data suggest that vascular EP4 receptors buffer the actions of AngII on renal hemodynamics and oxidative injury. CONCLUSION EP4 agonists may, therefore, protect against hypertension-associated kidney damage. Antioxid. Redox Signal. 25, 642-656.
Collapse
Affiliation(s)
- Jean-Francois Thibodeau
- 1 Chronic Disease Program, Department of Medicine, Kidney Research Centre, The Ottawa Hospital , Ottawa, Ontario, Canada .,2 Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa , Ontario, Canada
| | - Chet E Holterman
- 1 Chronic Disease Program, Department of Medicine, Kidney Research Centre, The Ottawa Hospital , Ottawa, Ontario, Canada
| | - Ying He
- 1 Chronic Disease Program, Department of Medicine, Kidney Research Centre, The Ottawa Hospital , Ottawa, Ontario, Canada
| | - Anthony Carter
- 2 Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa , Ontario, Canada
| | | | - Naomi C Boisvert
- 1 Chronic Disease Program, Department of Medicine, Kidney Research Centre, The Ottawa Hospital , Ottawa, Ontario, Canada .,2 Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa , Ontario, Canada
| | - Khaled S Abd-Elrahman
- 2 Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa , Ontario, Canada
| | - Karolynn J Hsu
- 2 Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa , Ontario, Canada
| | - Stephen S G Ferguson
- 2 Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa , Ontario, Canada
| | - Christopher R J Kennedy
- 1 Chronic Disease Program, Department of Medicine, Kidney Research Centre, The Ottawa Hospital , Ottawa, Ontario, Canada .,2 Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa , Ontario, Canada .,3 The Ottawa Hospital , Ottawa, Ontario, Canada
| |
Collapse
|
18
|
Poglitsch M, Sturrock ED, Danser AHJ. Letter to the editor: Angiotensin quantification by mass spectrometry. Am J Physiol Heart Circ Physiol 2016; 310:H452-3. [PMID: 26830339 DOI: 10.1152/ajpheart.00933.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
| | - E D Sturrock
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; and
| | - A H J Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
19
|
Sindone A, Erlich J, Lee C, Newman H, Suranyi M, Roger SD. Cardiovascular risk reduction in hypertension: angiotensin-converting enzyme inhibitors, angiotensin receptor blockers. Where are we up to? Intern Med J 2016; 46:364-72. [DOI: 10.1111/imj.12975] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 11/05/2015] [Accepted: 11/16/2015] [Indexed: 11/26/2022]
Affiliation(s)
- A. Sindone
- Heart Failure Unit and Department of Cardiac Rehabilitation; Concord Repatriation General Hospital; Concord New South Wales Australia
| | - J. Erlich
- Faculty of Medicine; University of NSW; Sydney New South Wales Australia
- Department of Nephrology; Prince of Wales Hospital; Sydney New South Wales Australia
| | - C. Lee
- Department of Cardiology; Nepean Hospital; Sydney New South Wales Australia
| | - H. Newman
- Department of Cardiology; Liverpool Hospital; Sydney New South Wales Australia
| | - M. Suranyi
- Liverpool Renal Clinical Research Centre; Liverpool Hospital; Sydney New South Wales Australia
| | - S. D. Roger
- Department of Renal Medicine; Gosford Hospital; Gosford New South Wales Australia
| |
Collapse
|
20
|
Affiliation(s)
- Mark C Chappell
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
21
|
Chappell MC. Biochemical evaluation of the renin-angiotensin system: the good, bad, and absolute? Am J Physiol Heart Circ Physiol 2015; 310:H137-52. [PMID: 26475588 DOI: 10.1152/ajpheart.00618.2015] [Citation(s) in RCA: 218] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 10/15/2015] [Indexed: 02/07/2023]
Abstract
The renin-angiotensin system (RAS) constitutes a key hormonal system in the physiological regulation of blood pressure through peripheral and central mechanisms. Indeed, dysregulation of the RAS is considered a major factor in the development of cardiovascular pathologies, and pharmacological blockade of this system by the inhibition of angiotensin-converting enzyme (ACE) or antagonism of the angiotensin type 1 receptor (AT1R) offers an effective therapeutic regimen. The RAS is now defined as a system composed of different angiotensin peptides with diverse biological actions mediated by distinct receptor subtypes. The classic RAS comprises the ACE-ANG II-AT1R axis that promotes vasoconstriction; water intake; sodium retention; and increased oxidative stress, fibrosis, cellular growth, and inflammation. In contrast, the nonclassical RAS composed primarily of the ANG II/ANG III-AT2R and the ACE2-ANG-(1-7)-AT7R pathways generally opposes the actions of a stimulated ANG II-AT1R axis. In lieu of the complex and multifunctional aspects of this system, as well as increased concerns on the reproducibility among laboratories, a critical assessment is provided on the current biochemical approaches to characterize and define the various components that ultimately reflect the status of the RAS.
Collapse
Affiliation(s)
- Mark C Chappell
- The Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
22
|
Sharp S, Poglitsch M, Zilla P, Davies NH, Sturrock ED. Pharmacodynamic effects of C-domain-specific ACE inhibitors on the renin-angiotensin system in myocardial infarcted rats. J Renin Angiotensin Aldosterone Syst 2015; 16:1149-58. [PMID: 25757657 DOI: 10.1177/1470320314568438] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 12/10/2014] [Indexed: 01/26/2023] Open
Abstract
INTRODUCTION The renin-angiotensin system (RAS) is a dynamic network that plays a critical role in blood pressure regulation and fluid and electrolyte homeostasis. Modulators of the RAS, such as angiotensin-converting enzyme (ACE) inhibitors, are widely used to treat hypertension, heart failure and myocardial infarction. METHODS The effect of ACE inhibitors (lisinopril and C-domain-selective LisW-S) on the constituent peptides of the RAS following myocardial infarction was examined in rats. Ten angiotensin peptides were analysed using a sensitive LC-MS/MS-based assay to examine both the circulating and equilibrium levels of these peptides. RESULTS Administration of lisinopril or LisW-S caused a significant decrease in Ang 1-8/Ang 1-10 ratios as determined by circulating and equilibrium peptide level analysis. Furthermore, Ang 1-7 levels were elevated by both ACE inhibitors, but only lisinopril decreased the Ang 1-5/Ang 1-7 ratio. This indicates LisW-S C-domain specificity as Ang 1-5 is generated by hydrolysis of Ang 1-7 by the N-domain. Further corroboration of LisW-S C-domain specificity is that only lisinopril increased the circulating levels of the N-domain ACE substrate Ac-SDKP. CONCLUSION LisW-S is able to effectively block ACE in vivo by C-domain-selective inhibition. The LC-MS/MS-based assay allows the evaluation of the pharmacologic impact of RAS inhibitors in different pathophysiological conditions.
Collapse
Affiliation(s)
- Sarah Sharp
- Cardiovascular Research Unit, Division of Cardiothoracic Surgery, University of Cape Town, South Africa
| | - Marko Poglitsch
- Attoquant Diagnostics GmbH, Campus-Vienna-Biocenter 5, Austria
| | - Peter Zilla
- Cardiovascular Research Unit, Division of Cardiothoracic Surgery, University of Cape Town, South Africa
| | - Neil H Davies
- Cardiovascular Research Unit, Division of Cardiothoracic Surgery, University of Cape Town, South Africa
| | - Edward D Sturrock
- Institute of Infectious Disease and Molecular Medicine, Division of Medical Biochemistry, University of Cape Town, South Africa
| |
Collapse
|
23
|
Thibodeau JF, Holterman CE, Burger D, Read NC, Reudelhuber TL, Kennedy CRJ. A novel mouse model of advanced diabetic kidney disease. PLoS One 2014; 9:e113459. [PMID: 25514595 PMCID: PMC4267730 DOI: 10.1371/journal.pone.0113459] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 10/27/2014] [Indexed: 01/11/2023] Open
Abstract
Currently available rodent models exhibit characteristics of early diabetic nephropathy (DN) such as hyperfiltration, mesangial expansion, and albuminuria yet features of late DN (hypertension, GFR decline, tubulointerstitial fibrosis) are absent or require a significant time investment for full phenotype development. Accordingly, the aim of the present study was to develop a mouse model of advanced DN with hypertension superimposed (HD mice). Mice transgenic for human renin cDNA under the control of the transthyretin promoter (TTRhRen) were employed as a model of angiotensin-dependent hypertension. Diabetes was induced in TTRhRen mice through low dose streptozotocin (HD-STZ mice) or by intercrossing with OVE26 diabetic mice (HD-OVE mice). Both HD-STZ and HD-OVE mice displayed more pronounced increases in urinary albumin levels as compared with their diabetic littermates. Additionally, HD mice displayed renal hypertrophy, advanced glomerular scarring and evidence of tubulointerstitial fibrosis. Both HD-OVE and HD-STZ mice showed evidence of GFR decline as FITC-inulin clearance was decreased compared to hyperfiltering STZ and OVE mice. Taken together our results suggest that HD mice represent a robust model of type I DN that recapitulates key features of human disease which may be significant in studying the pathogenesis of DN and in the assessment of putative therapeutics.
Collapse
Affiliation(s)
- Jean-Francois Thibodeau
- Kidney Research Centre, Division of Nephrology, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Chet E. Holterman
- Kidney Research Centre, Division of Nephrology, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Dylan Burger
- Kidney Research Centre, Division of Nephrology, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Naomi C. Read
- Kidney Research Centre, Division of Nephrology, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Timothy L. Reudelhuber
- Clinical Research Institute of Montreal, University of Montreal, Montreal, Quebec, Canada
| | - Christopher R. J. Kennedy
- Kidney Research Centre, Division of Nephrology, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- * E-mail:
| |
Collapse
|
24
|
Angiotensin converting enzyme is involved in the cardiac hypertrophy induced by sinoaortic denervation in rats. Cardiovasc Pathol 2014; 24:41-8. [PMID: 25261879 DOI: 10.1016/j.carpath.2014.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 08/11/2014] [Accepted: 08/25/2014] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION The present study was designed to test the hypothesis that local angiotensin converting enzyme (ACE) was involved in the cardiac hypertrophy induced by sinoaortic denervation (SAD) in rats. METHODS Experiment 1: Six weeks after SAD of rats, components of renin-angiotensin system (RAS) in left ventricles were assayed by quantitative real-time PCR and Western blotting analysis. Experiment 2: Rats were divided into five groups treated as follows: (1) sham-operated group; (2) SAD group; (3) SAD group treated with angiotensin II type 1 receptor (AT1R) antagonist losartan (10 mg·kg(-1)·day(-1), orally); (4) SAD group treated by ACE inhibitor ramipril (1 mg·kg(-1)·day(-1), orally); (5) SAD group treated by ramipril and the B2-kinin receptor selective antagonist HOE-140 (0.25 mg·kg(-1)·day(-1), subcutaneously). RESULTS SAD led to augmentation of the mRNA levels and protein expression of left ventricular ACE and AT1R. Both losartan and ramipril ameliorated SAD-induced left ventricular hypertrophy. Both losartan and ramipril abated oxidative stress, suppressed inflammation, and reduced expression TGFβ-R in left ventricles. In addition, the protective effect of ramipril could be abolished by HOE-140. CONCLUSION Local ACE is involved in the left ventricular hypertrophy induced by sinoaortic denervation in rats, via both angiotensin II/AT1R and bradykinin/B2R pathways.
Collapse
|