1
|
Thakur S, Mohanty P, Jadhav MS, Gaikwad AB, Jadhav HR. A perspective on the development of small molecular neprilysin inhibitors (NEPi) with emphasis on cardiorenal disease. Eur J Med Chem 2024; 280:116932. [PMID: 39378824 DOI: 10.1016/j.ejmech.2024.116932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/14/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024]
Abstract
Neprilysin is a cell surface metallo-endopeptidase, commonly identified as neutral endopeptidase (NEP), that plays a crucial role in the cleavage of peptides, for example, natriuretic peptides, angiotensin II, enkephalins, endothelin, bradykinin, substance P, glucagon-like peptide and amyloid beta. In the case of heart failure, a significant upsurge in NEP activity and expression enhances the degradation of natriuretic peptides. Therefore, NEP inhibitors have gained attention in the field of cardiology. NEP has been studied for over 40 years; however, it has recently gained attention with the US FDA approval of a fixed dose combination of sacubitril (NEP inhibitor) and valsartan (AT-1 inhibitor) for chronic heart failure treatment. The present review elucidates the role of neprilysin in cardiorenal disease, its pathophysiology, and how NEP inhibition benefits. It also summarizes the research advances in NEP inhibitors (NEPi) and their structure-activity relationships. Moreover, the review provides insight into NEPi effectiveness - alone or combined with other cardiorenal protective agents. It is expected to help medicinal chemists synthesize and develop novel NEPi.
Collapse
Affiliation(s)
- Shikha Thakur
- Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Pilani Campus, Vidya Vihar, Pilani, 333031, (RJ), India
| | - Priyanka Mohanty
- Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Pilani Campus, Vidya Vihar, Pilani, 333031, (RJ), India
| | - Madhav S Jadhav
- Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Pilani Campus, Vidya Vihar, Pilani, 333031, (RJ), India
| | - Anil Bhanudas Gaikwad
- Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Pilani Campus, Vidya Vihar, Pilani, 333031, (RJ), India
| | - Hemant R Jadhav
- Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Pilani Campus, Vidya Vihar, Pilani, 333031, (RJ), India.
| |
Collapse
|
2
|
Żukowska J, Moss SJ, Subramanian V, Acharya KR. Molecular basis of selective amyloid-β degrading enzymes in Alzheimer's disease. FEBS J 2024; 291:2999-3029. [PMID: 37622248 DOI: 10.1111/febs.16939] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/31/2023] [Accepted: 08/22/2023] [Indexed: 08/26/2023]
Abstract
The accumulation of the small 42-residue long peptide amyloid-β (Aβ) has been proposed as a major trigger for the development of Alzheimer's disease (AD). Within the brain, the concentration of Aβ peptide is tightly controlled through production and clearance mechanisms. Substantial experimental evidence now shows that reduced levels of Aβ clearance are present in individuals living with AD. This accumulation of Aβ can lead to the formation of large aggregated amyloid plaques-one of two detectable hallmarks of the disease. Aβ-degrading enzymes (ADEs) are major players in the clearance of Aβ. Stimulating ADE activity or expression, in order to compensate for the decreased clearance in the AD phenotype, provides a promising therapeutic target. It has been reported in mice that upregulation of ADEs can reduce the levels of Aβ peptide and amyloid plaques-in some cases, this led to improved cognitive function. Among several known ADEs, neprilysin (NEP), endothelin-converting enzyme-1 (ECE-1), insulin degrading enzyme (IDE) and angiotensin-1 converting enzyme (ACE) from the zinc metalloprotease family have been identified as important. These ADEs have the capacity to digest soluble Aβ which, in turn, cannot form the toxic oligomeric species. While they are known for their amyloid degradation, they exhibit complexity through promiscuous nature and a broad range of substrates that they can degrade. This review highlights current structural and functional understanding of these key ADEs, giving some insight into the molecular interactions that leads to the hydrolysis of peptide substrates, the crucial tasks performed by them and the potential for therapeutic use in the future.
Collapse
|
3
|
Banerjee S, Manisha C, Bharathi J J, Kumar AP, Justin A, Ramanathan M. Structural dynamics and catalytic modulations of Aβ regulating enzymes as future outlook for Alzheimer's. Biochem Biophys Res Commun 2022; 631:1-8. [PMID: 36162324 DOI: 10.1016/j.bbrc.2022.09.068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/15/2022] [Indexed: 11/28/2022]
Abstract
Aβ cascade hypothesis being considered most evident event in AD pathology and even today it holds good. Dysregulation of catalytic events of Aβ regulating enzymes can possibly cause faulty Aβ trafficking; inequity of Aβ formation and clearance resulting in misfolded protein accumulation, neurodegeneration and cognitive impairment. Many novel approaches have been made on this pathway to discover new molecules, unfortunately couldn't reach the terminal phases of clinical trials. Over decades, studies have been more focused on enzyme chemistry and explored the relationship between structural features and catalytic function of Aβ regulating enzymes. However, the modulations of catalytic mechanisms of those enzymes have not been imposed so far to reduce the Aβ load. Hence, in this review, we have critically detailed the knowledge of basic structural dynamics and possible catalytic modulations of enzymes responsible for Aβ formation and clearance that will impart new perspectives in drug discovery process.
Collapse
Affiliation(s)
- Sayani Banerjee
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, 643 001, India
| | - Chennu Manisha
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, 643 001, India
| | - Jeyaram Bharathi J
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, 643 001, India
| | - Ashwini Prem Kumar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, 643 001, India
| | - Antony Justin
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, 643 001, India.
| | - Muthiah Ramanathan
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, Tamil Nadu, 641 004, India
| |
Collapse
|
4
|
Lenz M, Krychtiuk KA, Brekalo M, Draxler DF, Pavo N, Hengstenberg C, Huber K, Hülsmann M, Heinz G, Wojta J, Speidl WS. Soluble neprilysin and survival in critically ill patients. ESC Heart Fail 2022; 9:1160-1166. [PMID: 35040286 PMCID: PMC8934932 DOI: 10.1002/ehf2.13787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/08/2021] [Accepted: 12/13/2021] [Indexed: 11/30/2022] Open
Abstract
Background Critically ill patients admitted to an intensive care unit (ICU) exhibit a high mortality rate irrespective of the initial cause of hospitalization. Neprilysin, a neutral endopeptidase degrading an array of vasoactive peptides became a drug target within the treatment of heart failure with reduced ejection fraction. The aim of this study was to analyse whether circulating levels of neprilysin at ICU admission are associated with 30 day mortality. Methods and results In this single‐centre prospective observational study, 222 consecutive patients admitted to a tertiary ICU at a university hospital were included. Blood was drawn at admission and soluble neprilysin levels were measured using ELISA. In the total cohort, soluble neprilysin levels did not differ according to survival status after 30 days as well as type of admission. However, in patients after surgery or heart valve intervention, 30 day survivors exhibited significantly lower circulating neprilysin levels as compared to those who died within 30 days (660.2, IQR: 156.4–2512.5 pg/mL vs. 6532.6, IQR: 1840.1–10 000.0 pg/mL; P = 0.02). Soluble neprilysin predicted mortality independently from age, gender, and commonly used scores of risk‐prediction (EuroSCORE II, STS‐score, and SAPS II score). Additionally, soluble neprilysin was markedly elevated in patients with sepsis and septic shock (P < 0.05). Conclusion At the time of ICU admission, circulating levels of neprilysin independently predicted 30 day mortality in patients following cardiac surgery or heart valve intervention, but not in critically ill medical patients. Furthermore, patients suffering from sepsis and septic shock displayed significantly increased circulating neprilysin levels.
Collapse
Affiliation(s)
- Max Lenz
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria.,Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria
| | - Konstantin A Krychtiuk
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria.,Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria
| | - Mira Brekalo
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Dominik F Draxler
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Noemi Pavo
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Christian Hengstenberg
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Kurt Huber
- Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria.,3rd Medical Department, Wilhelminenhospital, Vienna, Austria
| | - Martin Hülsmann
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Gottfried Heinz
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Johann Wojta
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria.,Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria.,Core Facilities, Medical University of Vienna, Vienna, Austria
| | - Walter S Speidl
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| |
Collapse
|
5
|
Choi H, Kim E, Choi JY, Park E, Lee HJ. Potent therapeutic targets for treatment of Alzheimer's disease: Amyloid degrading enzymes. B KOREAN CHEM SOC 2021. [DOI: 10.1002/bkcs.12390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Hang Choi
- Department of Chemistry Education Kongju National University Gongju Republic of Korea
| | - Eungchan Kim
- Department of Chemistry Education Kongju National University Gongju Republic of Korea
| | - Jae Yoon Choi
- Department of Chemistry Education Kongju National University Gongju Republic of Korea
| | - Eunsik Park
- Department of Life Sport Education Kongju National University Gongju Republic of Korea
| | - Hyuck Jin Lee
- Department of Chemistry Education Kongju National University Gongju Republic of Korea
| |
Collapse
|
6
|
Radaghdam S, Karamad V, Nourazarian A, Shademan B, Khaki-Khatibi F, Nikanfar M. Molecular mechanisms of sex hormones in the development and progression of Alzheimer's disease. Neurosci Lett 2021; 764:136221. [PMID: 34500000 DOI: 10.1016/j.neulet.2021.136221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/23/2021] [Accepted: 09/02/2021] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a form of brain disorder characterized by various pathological changes in the brain. Numerous studies have shown that sex hormones are involved in the disease. For instance, progesterone, estrogen, and testosterone are well-known steroid sex hormones that play an essential role in AD pathogenesis. The Gender-dependency of AD is attributed to the effect of these hormones on the brain, which plays a neuroprotective role. In recent years, much research has been performed on the protective role of these hormones against nerve cell damage, which are promising for AD management. Hence, in the current review, we aim to decipher the protective role of steroid hormones in AD. Accordingly, we will discuss their functional mechanisms at the genomic and non-genomic scales.
Collapse
Affiliation(s)
- Saeed Radaghdam
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahidreza Karamad
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Alireza Nourazarian
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Behrouz Shademan
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Fatemeh Khaki-Khatibi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Nikanfar
- Department of Neurology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
7
|
Leite JP, Lete MG, Fowler SB, Gimeno A, Rocha JF, Sousa SF, Webster CI, Jiménez-Bar̀bero JJ, Gales L. Aβ 31-35 Decreases Neprilysin-Mediated Alzheimer's Amyloid-β Peptide Degradation. ACS Chem Neurosci 2021; 12:3708-3718. [PMID: 34505762 DOI: 10.1021/acschemneuro.1c00432] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Alzheimer's disease is associated with the deposition of extracellular senile plaques, made primarily of amyloid-β (Aβ), particularly peptides Aβ1-42 and Aβ1-40. Neprilysin, or neutral endopeptidase (NEP), catalyzes proteolysis of the amyloid peptides (Aβ) and is recognized as one of the major regulators of the levels of these peptides in the brain, preventing Aβ accumulation and plaque formation. Here, we used a combination of techniques to elucidate the mechanism of Aβ binding and cleavage by NEP. Our findings indicate that the Aβ31-X cleavage products remain bound to the neprilysin active site, reducing proteolytic activity. Interestingly, it was already shown that this Aβ31-35 sequence is also critical for recognition of Aβ peptides by other targets, such as the serpin-enzyme complex receptor in neuronal cells.
Collapse
Affiliation(s)
- José P. Leite
- i3S—Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, Porto 4200-135, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, Porto 4200-135, Portugal
- Programa Doutoral em Biologia Molecular e Celular (MCbiology), ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Rua de Jorge Viterbo Ferreira 228, Porto 4050-313, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Marta G. Lete
- CIC bioGUNE, Bizkaia Technology Park, Building 801A, Derio 48170, Spain
| | - Susan B. Fowler
- Antibody Discovery & Protein Engineering R&D, AstraZeneca, Cambridge CB21 6GH, U.K
| | - Ana Gimeno
- CIC bioGUNE, Bizkaia Technology Park, Building 801A, Derio 48170, Spain
| | - Juliana F. Rocha
- UCIBIO/REQUIMTE, BioSIM-Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, Porto 4200-319, Portugal
| | - Sérgio F. Sousa
- UCIBIO/REQUIMTE, BioSIM-Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, Porto 4200-319, Portugal
| | - Carl I. Webster
- Antibody Discovery & Protein Engineering R&D, AstraZeneca, Cambridge CB21 6GH, U.K
| | - Jesús J. Jiménez-Bar̀bero
- CIC bioGUNE, Bizkaia Technology Park, Building 801A, Derio 48170, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao 48013, Spain
- Department of Organic Chemistry II, Faculty of Science and Technology, UPV-EHU, 48940 Leioa, Spain
| | - Luís Gales
- i3S—Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, Porto 4200-135, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, Porto 4200-135, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
8
|
Qu R, He K, Fan T, Yang Y, Mai L, Lian Z, Zhou Z, Peng Y, Khan AU, Sun B, Huang X, Ouyang J, Pan X, Dai J, Huang W. Single-cell transcriptomic sequencing analyses of cell heterogeneity during osteogenesis of human adipose-derived mesenchymal stem cells. STEM CELLS (DAYTON, OHIO) 2021; 39:1478-1488. [PMID: 34346140 DOI: 10.1002/stem.3442] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 07/19/2021] [Indexed: 11/05/2022]
Abstract
Mesenchymal stem cells (MSCs) are known for their multilineage differentiation potential with immune-modulatory properties. The molecular underpinnings of differentiation remain largely undefined. In this study, we investigated the cellular and molecular features of chemically induced osteogenesis from MSC isolated from human adipose tissue (human adipose MSCs, hAMSCs) using single-cell RNA-sequencing (scRNA-seq). We found that a near complete differentiation of osteogenic clusters from hAMSCs under a directional induction. Both groups of cells are heterogeneous, and some of the hAMSCs cells are intrinsically prepared for osteogenesis, while variant OS clusters seems in cooperation with a due division of the general function. We identified a set of genes related to cell stress response highly expressed during the differentiation. We also characterized a series of transitional transcriptional waves throughout the process from hAMSCs to osteoblast and specified the unique gene networks and epigenetic status as key markers of osteogenesis.
Collapse
Affiliation(s)
- Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Demonstration Center for Experimental Education of Basic Medical Sciences & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Kai He
- Guangdong Provincial Key Lab of Single Cell Technology and Application & Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, People's Republic of China
| | - Tingyu Fan
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Demonstration Center for Experimental Education of Basic Medical Sciences & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Yuchao Yang
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Demonstration Center for Experimental Education of Basic Medical Sciences & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Liyao Mai
- Guangdong Provincial Key Lab of Single Cell Technology and Application & Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, People's Republic of China
| | - Zhiwei Lian
- Guangdong Provincial Key Lab of Single Cell Technology and Application & Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, People's Republic of China
| | - Zhitao Zhou
- Central Laboratory, Southern Medical University, Guangzhou, People's Republic of China
| | - Yan Peng
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Demonstration Center for Experimental Education of Basic Medical Sciences & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Asmat Ullah Khan
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Demonstration Center for Experimental Education of Basic Medical Sciences & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Bing Sun
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Demonstration Center for Experimental Education of Basic Medical Sciences & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiaolan Huang
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Demonstration Center for Experimental Education of Basic Medical Sciences & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Demonstration Center for Experimental Education of Basic Medical Sciences & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Xinghua Pan
- Guangdong Provincial Key Lab of Single Cell Technology and Application & Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, People's Republic of China
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Demonstration Center for Experimental Education of Basic Medical Sciences & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Wenhua Huang
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Demonstration Center for Experimental Education of Basic Medical Sciences & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
9
|
Redox-Active Metal Ions and Amyloid-Degrading Enzymes in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22147697. [PMID: 34299316 PMCID: PMC8307724 DOI: 10.3390/ijms22147697] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/11/2021] [Accepted: 07/16/2021] [Indexed: 12/11/2022] Open
Abstract
Redox-active metal ions, Cu(I/II) and Fe(II/III), are essential biological molecules for the normal functioning of the brain, including oxidative metabolism, synaptic plasticity, myelination, and generation of neurotransmitters. Dyshomeostasis of these redox-active metal ions in the brain could cause Alzheimer’s disease (AD). Thus, regulating the levels of Cu(I/II) and Fe(II/III) is necessary for normal brain function. To control the amounts of metal ions in the brain and understand the involvement of Cu(I/II) and Fe(II/III) in the pathogenesis of AD, many chemical agents have been developed. In addition, since toxic aggregates of amyloid-β (Aβ) have been proposed as one of the major causes of the disease, the mechanism of clearing Aβ is also required to be investigated to reveal the etiology of AD clearly. Multiple metalloenzymes (e.g., neprilysin, insulin-degrading enzyme, and ADAM10) have been reported to have an important role in the degradation of Aβ in the brain. These amyloid degrading enzymes (ADE) could interact with redox-active metal ions and affect the pathogenesis of AD. In this review, we introduce and summarize the roles, distributions, and transportations of Cu(I/II) and Fe(II/III), along with previously invented chelators, and the structures and functions of ADE in the brain, as well as their interrelationships.
Collapse
|
10
|
Mascarello A, Azevedo H, Ferreira Junior MA, Ishikawa EE, Guimarães CRW. Design, synthesis and antihypertensive evaluation of novel codrugs with combined angiotensin type 1 receptor antagonism and neprilysin inhibition. Eur J Pharm Sci 2021; 159:105731. [PMID: 33493668 DOI: 10.1016/j.ejps.2021.105731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 10/22/2022]
Abstract
The multifactorial etiology of hypertension has promoted the research of blood pressure-lowering agents with multitarget actions to achieve better clinical outcomes. We describe here the discovery of novel dual-acting antihypertensive codrugs combining pharmacophores with angiotensin type 1 (AT1) receptor antagonism and neprilysin (NEP) inhibition. Specifically, the codrugs combine the AT1 antagonists losartan or its carboxylic acid active metabolite (E-3174) with selected monocarboxylic acid NEP inhibitors through a cleavable linker. The resulting codrugs exhibited high rates of in vitro conversion into the active molecules upon incubation with human/rat liver S9 fractions and in vivo conversion after oral administration in rodents. Moreover, the acute effects of one of the designed codrugs (3b) was confirmed at the doses of 10, 30 and 60 mg/kg p.o. in the spontaneous hypertensive rat (SHR) model, showing better antihypertensive response over 24 hours than the administration of an equivalent fixed-dose combination of 15 mg/kg of losartan and 14 mg/kg of the same NEP inhibitor used in 3b. The results demonstrate that the codrug approach is a plausible strategy to develop a single molecular entity with combined AT1 and NEP activities, aiming at achieving improved pharmacokinetics, efficacy and dosage convenience, as well as reduced drug-drug interaction for hypertension patients. In addition, the developability of the codrug should be comparable to the one of marketed AT1 antagonists, most of them prodrugs, but bearing only the AT1 pharmacophore.
Collapse
Affiliation(s)
| | - Hatylas Azevedo
- Aché Laboratórios Farmacêuticos, Guarulhos, São Paulo 07034-904, Brazil
| | | | | | | |
Collapse
|
11
|
Liang WG, Mancl JM, Zhao M, Tang WJ. Structural analysis of Mycobacterium tuberculosis M13 metalloprotease Zmp1 open states. Structure 2020; 29:709-720.e3. [PMID: 33378640 DOI: 10.1016/j.str.2020.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/12/2020] [Accepted: 12/04/2020] [Indexed: 12/16/2022]
Abstract
Zinc metalloprotease 1 (Zmp1), a Mycobacterium tuberculosis 75 kDa secreted enzyme, mediates key stages of tuberculosis disease progression. The biological activity of Zmp1 presumably stems from its ability to degrade bacterium- and/or host-derived peptides. The crystal structures of Zmp1 and related M13 metalloproteases, such as neprilysin and endothelin-converting enzyme-1 were determined only in the closed conformation, which cannot capture substrates or release proteolytic products. Thus, the mechanisms of substrate binding and selectivity remain elusive. Here we report two open-state cryo-EM structures of Zmp1, revealed by our SAXS analysis to be the dominant states in solution. Our structural analyses reveal how ligand binding induces a conformational switch in four linker regions to drive the rigid body motion of the D1 and D2 domains, which form the sizable catalytic chamber. Furthermore, they offer insights into the catalytic cycle and mechanism of substrate recognition of M13 metalloproteases for future therapeutic innovations.
Collapse
Affiliation(s)
- Wenguang G Liang
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Jordan M Mancl
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Minglei Zhao
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA.
| | - Wei-Jen Tang
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|