1
|
Forbes AK, Gallagher H. Initiating renin-angiotensin system inhibitors in chronic kidney disease. Drug Ther Bull 2025; 63:54-58. [PMID: 40174939 DOI: 10.1136/dtb.2023.000052] [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: 04/04/2025]
Abstract
Chronic kidney disease (CKD) is a heterogeneous group of disorders characterised by abnormalities in kidney structure or function, present for at least 3 months. Renin-angiotensin system (RAS) inhibitors are established treatments in the management of CKD, reducing the risk of adverse kidney and cardiovascular events. Despite an extensive evidence base and decades of clinical experience, they remain underused, underdosed and frequently discontinued due to concerns about changes in kidney function and hyperkalaemia. In this article, we provide an overview of initiating RAS inhibitors in adults with CKD, highlight current guideline recommendations and offer a practical approach to dealing with the challenges associated with their use.
Collapse
Affiliation(s)
- Anna Katharina Forbes
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Hugh Gallagher
- Renal Services, Epsom and Saint Helier University Hospitals NHS Trust, Carshalton, UK
- School of Primary Care, Population Sciences and Medical Education, University of Southampton, Southampton, UK
| |
Collapse
|
2
|
Zhou Y, Xu W, Ruan B, Zhu L, Jiang Y, Cai H, Huang J. Molecular Imaging of Renin Activity using Fluorogenic Nanoprobes for Precision Antihypertensive Therapy. Angew Chem Int Ed Engl 2025; 64:e202416002. [PMID: 39279688 DOI: 10.1002/anie.202416002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/18/2024]
Abstract
Life-threatening hypertension remains inadequately controlled in clinics due to its heterogeneous renin levels. Rapid stratification of hypertension through renin analysis is crucial for effective personalized treatment, yet an ultrasensitive detection approach is currently lacking. Here, we report activatable renin nanoprobes (ARNs) for non-invasive and ultrasensitive profiling of renin activity and guiding antihypertensive treatment decision through near-infrared fluorescence (NIRF) in vivo imaging and in vitro urinalysis. ARNs are intrinsically non-fluorescent due to NIRF reporter connected to a gold nanocluster through a renin-responsive peptide. In hyperreninemia mouse models, ARNs specifically react with renin to liberate the renal clearable NIRF reporter for accurate renin detection that outperforms the gold standard radioimmunoassay. Such specific and sensitive detection also enables imaging-based high-throughput screening of antihypertensive drugs. In hypertensive rat models, ARNs enable ultrasensitive detection of both plasma and urinary renin, facilitating renin-guided precision treatment and significantly improving hypertension control rate (90 % versus 58 %). Our nanoprobe platform holds great potential for assisting clinicians in rapidly and accurately classifying hypertensive patients and improving outcomes through tailored treatment selection.
Collapse
Affiliation(s)
- Ya Zhou
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key La-boratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Weiping Xu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key La-boratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Bankang Ruan
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key La-boratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Lijuan Zhu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key La-boratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yuyan Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA-94305, USA
| | - Hui Cai
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen, 518107, China
| | - Jiaguo Huang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key La-boratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| |
Collapse
|
3
|
Shen ZH, Ye T, Chen B, Wan C, Lu X, Chen TH, Lin S, Ye JX, Xie L, Fu YS. 5,7-Dihydroxyflavone acts on eNOS to achieve hypotensive effects in spontaneously hypertensive rats. Sci Rep 2025; 15:594. [PMID: 39747252 PMCID: PMC11695686 DOI: 10.1038/s41598-024-84259-6] [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: 09/22/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
Hypertension is one of the most serious chronic diseases. This study will focus on the systemic antihypertensive mechanisms of 5,7-dihydroxyflavone from in silico simulations to in vivo validations. In-silico studies were applied by network pharmacology, molecular docking, and molecular dynamic simulation. Based on the information of network pharmacology, 5,7-dihydroxyflavone could act on several different blood pressure regulating pathways, molecular docking results confirmed it might direct binding on the active pocket of eNOS, and the average molecular distance between 5,7-dihydroxyflavone -eNOS is less than 0.4 Å by molecular dynamic simulation. The in vivo studies were carried by SHRs oral administrated with 10 mg/kg 5,7-dihydroxyflavone that could alleviate hypertensive symptoms within 30 min, but if SHRs pretreated with L-NAME (10 mg/kg, an eNOS inhibitor) can erase the anti-hypertensive effects of 5,7-dihydroxyflavone, but no affected by aminoguanidine pretreatment (100 mg/kg, the selective antagonist of iNOS). Furthermore, oral administration of 5,7-dihydroxyflavone does not affect the heart rate and pulse pressure difference in SHR rats. In conclusion, the effects of 5,7-dihydroxyflavone on blood pressure regulation may act on eNOS as an agonist to achieve its acute antihypertensive effects. These acute antihypertensive effects suggest that 5,7-dihydroxyflavone has the potential to be a candidate medication for urgently lowering blood pressure requirements without posing hypertensive risks.
Collapse
Affiliation(s)
- Zi-Han Shen
- College of Clinical Medicine, Xiamen Medical College, Xiamen, Fujian, China
| | - Tongjie Ye
- College of Clinical Medicine, Xiamen Medical College, Xiamen, Fujian, China
| | - Baozhen Chen
- College of Pharmacy, Xiamen Medical College, Xiamen, Fujian, China
| | - Congchao Wan
- College of Clinical Medicine, Xiamen Medical College, Xiamen, Fujian, China
| | - Xiaolin Lu
- Department of Neurology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian, China
| | - Ting-Hsu Chen
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Shuang Lin
- College of Clinical Medicine, Xiamen Medical College, Xiamen, Fujian, China
| | - Jia-Xin Ye
- College of Clinical Medicine, Xiamen Medical College, Xiamen, Fujian, China
| | - Liping Xie
- Anatomy Department, College of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, China
| | - Yaw-Syan Fu
- Anatomy Department, College of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, China.
- Institute of Respiratory Disease, College of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, China.
| |
Collapse
|
4
|
Martin PJP, Willicombe M, Roufosse C. Angiotensin II Type-1 Receptor Antibody in Solid Organ Transplantation - Is It Time to Test? Transpl Int 2024; 37:13280. [PMID: 39606688 PMCID: PMC11598415 DOI: 10.3389/ti.2024.13280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/15/2024] [Indexed: 11/29/2024]
Abstract
Angiotensin II type-1 receptor antibody (AT1R-Ab) has been mooted as a potential effector of both acute and chronic antibody mediated rejection (AMR). A growing body of literature on the topic is now coming under scrutiny in the context of the evolving Banff AMR diagnostic classification system and refinement of recommendations for histocompatibility testing by the Sensitization in Transplantation Assessment of Risk (STAR) workgroup. This mini-review discusses the latest understanding of pathophysiological mechanisms, clinical evidence for the pathogenicity of AT1R-Ab, and methods of laboratory testing.
Collapse
Affiliation(s)
- Paul James Patrick Martin
- Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, United Kingdom
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom
| | - Michelle Willicombe
- Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, United Kingdom
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom
| | - Candice Roufosse
- Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, United Kingdom
- Department of Histopathology, Northwest London Pathology NHS Trust, Charing Cross Hospital, London, United Kingdom
| |
Collapse
|
5
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Overview of pyroptosis mechanism and in-depth analysis of cardiomyocyte pyroptosis mediated by NF-κB pathway in heart failure. Biomed Pharmacother 2024; 179:117367. [PMID: 39214011 DOI: 10.1016/j.biopha.2024.117367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
The pyroptosis of cardiomyocytes has become an essential topic in heart failure research. The abnormal accumulation of these biological factors, including angiotensin II, advanced glycation end products, and various growth factors (such as connective tissue growth factor, vascular endothelial growth factor, transforming growth factor beta, among others), activates the nuclear factor-κB (NF-κB) signaling pathway in cardiovascular diseases, ultimately leading to pyroptosis of cardiomyocytes. Therefore, exploring the underlying molecular biological mechanisms is essential for developing novel drugs and therapeutic strategies. However, our current understanding of the precise regulatory mechanism of this complex signaling pathway in cardiomyocyte pyroptosis is still limited. Given this, this study reviews the milestone discoveries in the field of pyroptosis research since 1986, analyzes in detail the similarities, differences, and interactions between pyroptosis and other cell death modes (such as apoptosis, necroptosis, autophagy, and ferroptosis), and explores the deep connection between pyroptosis and heart failure. At the same time, it depicts in detail the complete pathway of the activation, transmission, and eventual cardiomyocyte pyroptosis of the NF-κB signaling pathway in the process of heart failure. In addition, the study also systematically summarizes various therapeutic approaches that can inhibit NF-κB to reduce cardiomyocyte pyroptosis, including drugs, natural compounds, small molecule inhibitors, gene editing, and other cutting-edge technologies, aiming to provide solid scientific support and new research perspectives for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
6
|
Liang Z, Wang J, Zhang H, Gao L, Xu J, Li P, Yang J, Fu X, Duan H, Liu J, Liu T, Ma W, Wu K. Peptide S4 is an entry inhibitor of SARS-CoV-2 infection. Virology 2024; 597:110149. [PMID: 38917689 DOI: 10.1016/j.virol.2024.110149] [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: 03/11/2024] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a significant socioeconomic burden, and combating COVID-19 is imperative. Blocking the SARS-CoV-2 RBD-ACE2 interaction is a promising therapeutic approach for viral infections, as SARS-CoV-2 binds to the ACE2 receptors of host cells via the RBD of spike proteins to infiltrate these cells. We used computer-aided drug design technology and cellular experiments to screen for peptide S4 with high affinity and specificity for the human ACE2 receptor through structural analysis of SARS-CoV-2 and ACE2 interactions. Cellular experiments revealed that peptide S4 effectively inhibited SARS-CoV-2 and HCoV-NL63 viruses from infecting host cells and was safe for cells at effective concentrations. Based on these findings, peptide S4 may be a potential pharmaceutical agent for clinical application in the treatment of the ongoing SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Zhiyu Liang
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China; Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jiamei Wang
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Huan Zhang
- Guangdong Center for Disease Control and Prevention, Guangdong, China
| | - Lixia Gao
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jun Xu
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Peiran Li
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jie Yang
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Xinting Fu
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Han Duan
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jiayan Liu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China; Institute of Antibody Engineering, School of Laboratory Medicine & Biotechnology, Southern Medical University, Guangzhou, China
| | - Tiancai Liu
- Institute of Antibody Engineering, School of Laboratory Medicine & Biotechnology, Southern Medical University, Guangzhou, China
| | - Weifeng Ma
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| | - Kun Wu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
7
|
Samsamikor M, Mackay DS, Mollard RC, Alashi AM, Aluko RE. Hemp seed protein and its hydrolysate compared with casein protein consumption in adults with hypertension: a double-blind crossover study. Am J Clin Nutr 2024; 120:56-65. [PMID: 38710445 PMCID: PMC11251217 DOI: 10.1016/j.ajcnut.2024.05.001] [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: 03/10/2023] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND The effects of consuming hemp seed protein (HSP) as well as its hydrolysate-derived bioactive peptide (HSP+) on blood pressure (BP) has not, to our knowledge, been investigated in humans. OBJECTIVES We aimed to investigate how consumption of HSP and its hydrolysate modulates 24-h systolic (SBP) and diastolic BP (DBP) and plasma biomarkers of BP compared with casein. METHODS In a double-blind, randomized, crossover design trial, 35 adults who had mild hypertension with SBP between 130 and 160 mmHg and DBP ≤110 mmHg were recruited. Participants were randomly assigned to varying sequences of 3 6-wk treatments, 50 g casein/d, 50 g HSP/d, or 45 g HSP plus 5 g HSP-derived bioactive peptides/d (HSP+), separated by a 2-wk washout period. Treatment effects were assessed with a linear mixed model with repeated measures. RESULTS Compared with casein, after HSP+ consumption, 24-h SBP and 24-h DBP decreased from 135.1 and 80.0 mmHg to 128.1 ± 1.6 (P < 0.0001) and 76.0 ± 1.4 mmHg (P < 0.0001), respectively, whereas these values were 133.5 ± 1.6 and 78.9 ± 1.4 mmHg after HSP consumption (P < 0.0001). There were no differences between the HSP and HSP+ consumption in plasma angiotensin-converting enzyme (ACE) activity, renin, or nitric oxide (NO) concentrations. However, these 2 treatments were able to lower both ACE and renin activities and raise NO concentration in plasma compared with casein. CONCLUSIONS These results suggest that hemp protein consumption, as well as in combination with bioactive peptides, may have a role in the dietary management of hypertension. This trial was registered at clinicaltrials.gov as NCT03508895.
Collapse
Affiliation(s)
- Maryam Samsamikor
- Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Dylan S Mackay
- Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Community Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Rebecca C Mollard
- Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Chronic Disease Innovation Centre, Seven Oaks General Hospital, Winnipeg, MB, Canada
| | - Adeola M Alashi
- Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Rotimi E Aluko
- Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Richardson Centre for Food Technology and Research, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
8
|
Shen Y, Lotenberg K, Zaworski J, Broeker KAE, Vasseur F, Louedec L, Placier S, Frère P, Verpont MC, Galichon P, Buob D, Hadchouel J, Terzi F, Chatziantoniou C, Calmont A. Neuropilin-1 regulates renin synthesis in juxtaglomerular cells. J Physiol 2024; 602:1815-1833. [PMID: 38381008 DOI: 10.1113/jp285422] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/06/2024] [Indexed: 02/22/2024] Open
Abstract
Renin is the key enzyme of the systemic renin-angiotensin-aldosterone system, which plays an essential role in regulating blood pressure and maintaining electrolyte and extracellular volume homeostasis. Renin is mainly produced and secreted by specialized juxtaglomerular (JG) cells in the kidney. In the present study, we report for the first time that the conserved transmembrane receptor neuropilin-1 (NRP1) participates in the development of JG cells and plays a key role in renin production. We used the myelin protein zero-Cre (P0-Cre) to abrogate Nrp1 constitutively in P0-Cre lineage-labelled cells of the kidney. We found that the P0-Cre precursor cells differentiate into renin-producing JG cells. We employed a lineage-tracing strategy combined with RNAscope quantification and metabolic studies to reveal a cell-autonomous role for NRP1 in JG cell function. Nrp1-deficient animals displayed abnormal levels of tissue renin expression and failed to adapt properly to a homeostatic challenge to sodium balance. These findings provide new insights into cell fate decisions and cellular plasticity operating in P0-Cre-expressing precursors and identify NRP1 as a novel key regulator of JG cell maturation. KEY POINTS: Renin is a centrepiece of the renin-angiotensin-aldosterone system and is produced by specialized juxtaglomerular cells (JG) of the kidney. Neuropilin-1 (NRP1) is a conserved membrane-bound receptor that regulates vascular and neuronal development, cancer aggressiveness and fibrosis progression. We used conditional mutagenesis and lineage tracing to show that NRP1 is expressed in JG cells where it regulates their function. Cell-specific Nrp1 knockout mice present with renin paucity in JG cells and struggle to adapt to a homeostatic challenge to sodium balance. The results support the versatility of renin-producing cells in the kidney and may open new avenues for therapeutic approaches.
Collapse
Affiliation(s)
- Yunzhu Shen
- Sorbonne Université, INSERM, Unité mixte de Recherche 1155, Kidney Research Centre, Hôpital Tenon, Paris, France
| | - Kenza Lotenberg
- Sorbonne Université, INSERM, Unité mixte de Recherche 1155, Kidney Research Centre, Hôpital Tenon, Paris, France
| | - Jeremy Zaworski
- Sorbonne Université, INSERM, Unité mixte de Recherche 1155, Kidney Research Centre, Hôpital Tenon, Paris, France
| | | | - Florence Vasseur
- Institut Necker Enfants Malades, Growth and Signalling departement, Université Paris Cité, INSERM U1151, CNRS UMR 8253, Paris, France
| | - Liliane Louedec
- Sorbonne Université, INSERM, Unité mixte de Recherche 1155, Kidney Research Centre, Hôpital Tenon, Paris, France
| | - Sandrine Placier
- Sorbonne Université, INSERM, Unité mixte de Recherche 1155, Kidney Research Centre, Hôpital Tenon, Paris, France
| | - Perrine Frère
- Sorbonne Université, INSERM, Unité mixte de Recherche 1155, Kidney Research Centre, Hôpital Tenon, Paris, France
| | - Marie-Christine Verpont
- Sorbonne Université, INSERM, Unité mixte de Recherche 1155, Kidney Research Centre, Hôpital Tenon, Paris, France
| | - Pierre Galichon
- Sorbonne Université, INSERM, Unité mixte de Recherche 1155, Kidney Research Centre, Hôpital Tenon, Paris, France
| | - David Buob
- Sorbonne Université, INSERM, Unité mixte de Recherche 1155, Kidney Research Centre, Hôpital Tenon, Paris, France
| | - Juliette Hadchouel
- Sorbonne Université, INSERM, Unité mixte de Recherche 1155, Kidney Research Centre, Hôpital Tenon, Paris, France
| | - Fabiola Terzi
- Institut Necker Enfants Malades, Growth and Signalling departement, Université Paris Cité, INSERM U1151, CNRS UMR 8253, Paris, France
| | - Christos Chatziantoniou
- Sorbonne Université, INSERM, Unité mixte de Recherche 1155, Kidney Research Centre, Hôpital Tenon, Paris, France
| | - Amélie Calmont
- Sorbonne Université, INSERM, Unité mixte de Recherche 1155, Kidney Research Centre, Hôpital Tenon, Paris, France
| |
Collapse
|
9
|
Bhandari S, Mehta S, Khawaja A, Cleland JGF, Ives N, Cockwell P. Evaluation of the stopping angiotensin converting enzyme inhibitor compared to angiotensin receptor blocker (STOP ACEi trial) in advanced and progressive chronic kidney disease. Kidney Int 2024; 105:200-208. [PMID: 37783444 DOI: 10.1016/j.kint.2023.09.012] [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: 05/30/2023] [Revised: 09/06/2023] [Accepted: 09/15/2023] [Indexed: 10/04/2023]
Abstract
In the STOP-ACEi trial, the outcome was similar whether or not renin-angiotensin system inhibitors (RASi) were discontinued. We now investigate whether the effect of withdrawing angiotensin converting enzyme inhibitors (ACEi) or angiotensin-receptor blockers (ARBs) differed. In this open label trial patients with estimated glomerular filtration rates (eGFR) under 30ml/min per 1.73 m2 and progressive chronic kidney disease (CKD) were randomized to stop or continue RASi. The primary outcome was eGFR at three years. The composite of kidney failure, over 50% fall in eGFR, or kidney replacement therapy (KRT) was also assessed. Of patients randomized, 99 stopped and 123 patients continued ACEi while 104 stopped and 77 continued ARB at baseline. At three years, the eGFR was similar whether or not patients were withdrawn from ACEi or from ARB. Kidney failure or initiation of KRT occurred in 65% of those stopping and 54% continuing ACEi (hazard ratio if stopped, 1.52; 95% Confidence Interval, 1.07 to 2.16) and in 60% on an ARB regardless of randomized group (hazard ratio if stopped, 1.23; 0.83 to 1.81). Kidney failure/Initiation of KRT with over 50% decline in eGFR occurred in 71% of those stopping and 59% continuing ACEi (relative risk if stopped, 1.19; 95% CI, 1.00 to 1.41) and in 65% stopping and 69% continuing ARB (relative risk if stopped, 0.96; 0.79 to 1.16). Thus, neither discontinuing ACEi nor ARB slowed the rate of decline in eGFR. Although discontinuation of ACEi appeared to have more unfavorable effects on kidney outcomes than stopping ARB, the trial was neither designed nor powered to show differences between agents.
Collapse
Affiliation(s)
- Sunil Bhandari
- Department of Renal Medicine, Hull University Teaching Hospitals NHS Trust, and Hull York Medical School, East Yorkshire, UK.
| | - Samir Mehta
- Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Arif Khawaja
- Department of Nephrology, Sheffield Kidney Institute, Sheffield, UK
| | - John G F Cleland
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Natalie Ives
- Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Paul Cockwell
- Department of Renal Medicine, Queen Elizabeth Hospital, Birmingham, UK
| |
Collapse
|
10
|
Omidkhah N, Hadizadeh F, Ghodsi R, Kesharwani P, Sahebkar A. In silico Evaluation of NO-Sartans against SARS-CoV-2. Curr Drug Discov Technol 2024; 21:e050324227669. [PMID: 38445698 DOI: 10.2174/0115701638279362240223070810] [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: 10/20/2023] [Revised: 01/16/2024] [Accepted: 01/29/2024] [Indexed: 03/07/2024]
Abstract
INTRODUCTION Numerous clinical trials are currently investigating the potential of nitric oxide (NO) as an antiviral agent against coronaviruses, including SARS-CoV-2. Additionally, some researchers have reported positive effects of certain Sartans against SARS-CoV-2. METHOD Considering the impact of NO-Sartans on the cardiovascular system, we have compiled information on the general structure, synthesis methods, and biological studies of synthesized NOSartans. In silico evaluation of all NO-Sartans and approved sartans against three key SARS-CoV- -2 targets, namely Mpro (PDB ID: 6LU7), NSP16 (PDB ID: 6WKQ), and ACE-2 (PDB ID: 1R4L), was performed using MOE. RESULTS Almost all NO-Sartans and approved sartans demonstrated promising results in inhibiting these SARS-CoV-2 targets. Compound 36 (CLC-1280) showed the best docking scores against the three evaluated targets and was further evaluated using molecular dynamics (MD) simulations. CONCLUSION Based on our in silico studies, CLC-1280 (a Valsartan dinitrate) has the potential to be considered as an inhibitor of the SARS-CoV-2 virus. However, further in vitro and in vivo evaluations are necessary for the drug development process.
Collapse
Affiliation(s)
- Negar Omidkhah
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Hadizadeh
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Razieh Ghodsi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi, 110062, India
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Hanif A, Edin ML, Zeldin DC, Nayeem MA. Overexpression of Human Soluble Epoxide Hydrolase Exacerbates Coronary Reactive Hyperemia Reduction in Angiotensin-II-Treated Mouse Hearts. J Cardiovasc Pharmacol 2024; 83:46-54. [PMID: 37788350 PMCID: PMC10841723 DOI: 10.1097/fjc.0000000000001490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/21/2023] [Indexed: 10/05/2023]
Abstract
ABSTRACT Coronary reactive hyperemia (CRH) is impaired in cardiovascular diseases, and angiotensin-II (Ang-II) exacerbates it. However, it is unknown how Ang-II affects CRH in Tie2-sEH Tr (human-sEH-overexpressed) versus wild-type (WT) mice. sEH-overexpression resulted in CRH reduction in Tie2-sEH Tr versus WT. We hypothesized that Ang-II exacerbates CRH reduction in Tie2-sEH Tr versus WT. The Langendorff system measured coronary flow in Tie2-sEH Tr and WT. The hearts were exposed to 15-second ischemia, and CRH was assessed in 10 mice each. Repayment volume was reduced by 40.50% in WT treated with Ang-II versus WT (7.42 ± 0.8 to 4.49 ± 0.8 mL/g) and 48% in Tie2-sEH Tr treated with Ang-II versus Tie2-sEH Tr (5.18 ± 0.4 to 2.68 ± 0.3 mL/g). Ang-II decreased repayment duration by 50% in WT-treated with Ang-II versus WT (2.46 ± 0.5 to 1.24 ± 0.4 minutes) and 54% in Tie2-sEH Tr treated with Ang-II versus Tie2-sEH Tr (1.66 ± 0.4 to 0.76 ± 0.2 minutes). Peak repayment flow was reduced by 11.2% in WT treated with Ang-II versus WT (35.98 ± 0.7 to 32.11 ± 1.4 mL/g) and 4% in Tie2-sEH Tr treated with Ang-II versus Tie2-sEH Tr (32.18 ± 0.6 to 30.89 ± 1.5 mL/g). Furthermore, coronary flow was reduced by 43% in WT treated with Ang-II versus WT (14.2 ± 0.5 to 8.15 ± 0.8 mL/min/g) and 32% in Tie2-sEH Tr treated with Ang-II versus Tie2-sEH Tr (12.1 ± 0.8 to 8.3 ± 1.2 mL/min/g). Moreover, the Ang-II-AT 1 -receptor and CYP4A were increased in Tie2-sEHTr. Our results demonstrate that Ang-II exacerbates CRH reduction in Tie2-sEH Tr mice.
Collapse
Affiliation(s)
- Ahmad Hanif
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Matthew L. Edin
- Division of Intramural Research, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Darryl C. Zeldin
- Division of Intramural Research, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Mohammed A. Nayeem
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
12
|
Gayathiri E, Prakash P, Kumaravel P, Jayaprakash J, Ragunathan MG, Sankar S, Pandiaraj S, Thirumalaivasan N, Thiruvengadam M, Govindasamy R. Computational approaches for modeling and structural design of biological systems: A comprehensive review. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 185:17-32. [PMID: 37821048 DOI: 10.1016/j.pbiomolbio.2023.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 08/14/2023] [Accepted: 08/27/2023] [Indexed: 10/13/2023]
Abstract
The convergence of biology and computational science has ushered in a revolutionary era, revolutionizing our understanding of biological systems and providing novel solutions to global problems. The field of genetic engineering has facilitated the manipulation of genetic codes, thus providing opportunities for the advancement of innovative disease therapies and environmental enhancements. The emergence of bio-molecular simulation represents a significant advancement in this particular field, as it offers the ability to gain microscopic insights into molecular-level biological processes over extended periods. Biomolecular simulation plays a crucial role in advancing our comprehension of organismal mechanisms by establishing connections between molecular structures, interactions, and biological functions. The field of computational biology has demonstrated its significance in deciphering intricate biological enigmas through the utilization of mathematical models and algorithms. The process of decoding the human genome has resulted in the advancement of therapies for a wide range of genetic disorders, while the simulation of biological systems contributes to the identification of novel pharmaceutical compounds. The potential of biomolecular simulation and computational biology is vast and limitless. As the exploration of the underlying principles that govern living organisms progresses, the potential impact of this understanding on cancer treatment, environmental restoration, and other domains is anticipated to be transformative. This review examines the notable advancements achieved in the field of computational biology, emphasizing its potential to revolutionize the comprehension and enhancement of biological systems.
Collapse
Affiliation(s)
- Ekambaram Gayathiri
- Department of Plant Biology and Plant Biotechnology, Guru Nanak College (Autonomous), Chennai, 42, Tamil Nadu, India
| | - Palanisamy Prakash
- Department of Botany, Periyar University, Periyar Palkalai Nagar, Salem, 636011, Tamil Nadu, India
| | - Priya Kumaravel
- Department of Biotechnology, St. Joseph College (Arts & Science), Kovur, Chennai, Tamil Nadu, India
| | - Jayanthi Jayaprakash
- Department of Advanced Zoology and Biotechnology, Guru Nanak College, Chennai, Tamil Nadu, India
| | | | - Sharmila Sankar
- Department of Advanced Zoology and Biotechnology, Guru Nanak College, Chennai, Tamil Nadu, India
| | - Saravanan Pandiaraj
- Department of Self-Development Skills, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Natesan Thirumalaivasan
- Department of Periodontics, Saveetha Dental College, and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMTAS), Chennai, 600077, Tamil Nadu, India
| | - Muthu Thiruvengadam
- Department of Applied Bioscience, College of Life and Environmental Sciences, Konkuk University, Seoul, 05029, South Korea
| | - Rajakumar Govindasamy
- Department of Orthodontics, Saveetha Dental College and Hospitals, Saveetha University, Chennai, India.
| |
Collapse
|
13
|
Forero K, Buqaileh R, Sunderman C, AbouAlaiwi W. COVID-19 and Neurological Manifestations. Brain Sci 2023; 13:1137. [PMID: 37626493 PMCID: PMC10452375 DOI: 10.3390/brainsci13081137] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a newly emerged coronavirus, has sparked a global pandemic with its airborne transmission and ability to infect with asymptomatic patients. The pathophysiology is thought to relate to the binding of angiotensin converting enzyme 2 (ACE2) receptors in the body. These receptors are widely expressed in various body organs such as the lungs, the heart, the gastrointestinal tract (GIT), and the brain. This article reviews the current knowledge on the symptoms of coronavirus disease 2019 (COVID-19), highlighting the neurological symptoms that are associated with COVID-19, and discussing the possible mechanisms for SARS-CoV-2 virus infection in the body.
Collapse
Affiliation(s)
| | | | | | - Wissam AbouAlaiwi
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA; (K.F.); (R.B.); (C.S.)
| |
Collapse
|
14
|
Nguyen BY, Zhou F, Binder P, Liu W, Hille SS, Luo X, Zi M, Zhang H, Adamson A, Ahmed FZ, Butterworth S, Cartwright EJ, Müller OJ, Guan K, Fitzgerald EM, Wang X. Prolylcarboxypeptidase Alleviates Hypertensive Cardiac Remodeling by Regulating Myocardial Tissue Angiotensin II. J Am Heart Assoc 2023; 12:e028298. [PMID: 37318028 PMCID: PMC10356030 DOI: 10.1161/jaha.122.028298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 05/25/2023] [Indexed: 06/16/2023]
Abstract
Background Prolonged activation of angiotensin II is the main mediator that contributes to the development of heart diseases, so converting angiotensin II into angiotensin 1-7 has emerged as a new strategy to attenuate detrimental effects of angiotensin II. Prolylcarboxypeptidase is a lysosomal pro-X carboxypeptidase that is able to cleave angiotensin II at a preferential acidic pH optimum. However, insufficient attention has been given to the cardioprotective functions of prolylcarboxylpeptidase. Methods and Results We established a CRISPR/CRISPR-associated protein 9-mediated global prolylcarboxylpeptidase-knockout and adeno-associated virus serotype 9-mediated cardiac prolylcarboxylpeptidase overexpression mouse models, which were challenged with the angiotensin II infusion (2 mg/kg per day) for 4 weeks, aiming to investigate the cardioprotective effect of prolylcarboxylpeptidase against hypertensive cardiac hypertrophy. Prolylcarboxylpeptidase expression was upregulated after 2 weeks of angiotensin II infusion and then became downregulated afterward in wild-type mouse myocardium, suggesting its compensatory function against angiotensin II stress. Moreover, angiotensin II-treated prolylcarboxylpeptidase-knockout mice showed aggravated cardiac remodeling and dampened cardiac contractility independent of hypertension. We also found that prolylcarboxylpeptidase localizes in cardiomyocyte lysosomes, and loss of prolylcarboxylpeptidase led to excessive angiotensin II levels in myocardial tissue. Further screening demonstrated that hypertrophic prolylcarboxylpeptidase-knockout hearts showed upregulated extracellular signal-regulated kinases 1/2 and downregulated protein kinase B activities. Importantly, adeno-associated virus serotype 9-mediated restoration of prolylcarboxylpeptidase expression in prolylcarboxylpeptidase-knockout hearts alleviated angiotensin II-induced hypertrophy, fibrosis, and cell death. Interestingly, the combination of adeno-associated virus serotype 9-mediated prolylcarboxylpeptidase overexpression and an antihypertensive drug, losartan, likely conferred more effective protection than a single treatment protocol to mitigate angiotensin II-induced cardiac dysfunction. Conclusions Our data demonstrate that prolylcarboxylpeptidase protects the heart from angiotensin II-induced hypertrophic remodeling by controlling myocardial angiotensin II levels.
Collapse
Affiliation(s)
- Binh Y. Nguyen
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Fangchao Zhou
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Pablo Binder
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Wei Liu
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Susanne S. Hille
- Department of Internal Medicine IIIUniversity of KielKielGermany
| | - Xiaojing Luo
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav CarusTechnische Universitaet DresdenDresdenGermany
| | - Min Zi
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Hongyuan Zhang
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Antony Adamson
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Fozia Z. Ahmed
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Sam Butterworth
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | | | - Oliver J. Müller
- Department of Internal Medicine IIIUniversity of KielKielGermany
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav CarusTechnische Universitaet DresdenDresdenGermany
| | | | - Xin Wang
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| |
Collapse
|
15
|
Zhang X, Zhang S, Wang M, Chen H, Liu H. Advances in the allostery of angiotensin II type 1 receptor. Cell Biosci 2023; 13:110. [PMID: 37330563 DOI: 10.1186/s13578-023-01063-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 05/31/2023] [Indexed: 06/19/2023] Open
Abstract
Angiotensin II type 1 receptor (AT1R) is a promising therapeutic target for cardiovascular diseases. Compared with orthosteric ligands, allosteric modulators attract considerable attention for drug development due to their unique advantages of high selectivity and safety. However, no allosteric modulators of AT1R have been applied in clinical trials up to now. Except for the classical allosteric modulators of AT1R such as antibody, peptides and amino acids, cholesterol and biased allosteric modulators, there are non-classical allosteric modes including the ligand-independent allosteric mode, and allosteric mode of biased agonists and dimers. In addition, finding the allosteric pockets based on AT1R conformational change and interaction interface of dimers are the future of drug design. In this review, we summarize the different allosteric mode of AT1R, with a view to contribute to the development and utilization of drugs targeting AT1R allostery.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Suli Zhang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Meili Wang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Hao Chen
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Huirong Liu
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China.
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, People's Republic of China.
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 10 Xitoutiao, You An Men Street, Beijing, 100069, China.
| |
Collapse
|
16
|
Paidi RK, Jana M, Raha S, Mishra RK, Jeong B, Sheinin M, Pahan K. Prenol, but Not Vitamin C, of Fruit Binds to SARS-CoV-2 Spike S1 to Inhibit Viral Entry: Implications for COVID-19. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1938-1949. [PMID: 37144841 PMCID: PMC10615733 DOI: 10.4049/jimmunol.2200279] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 02/23/2023] [Indexed: 05/06/2023]
Abstract
Fruit consumption may be beneficial for fighting infection. Although vitamin C is the celebrity component of fruit, its role in COVID-19 is unclear. Because spike S1 of SARS-CoV-2 binds to angiotensin-converting enzyme 2 (ACE2) on host cells to enter the cell and initiate COVID-19, using an α-screen-based assay, we screened vitamin C and other components of fruit for inhibiting the interaction between spike S1 and ACE2. We found that prenol, but neither vitamin C nor other major components of fruit (e.g., cyanidin and rutin), reduced the interaction between spike S1 and ACE2. Thermal shift assays indicated that prenol associated with spike S1, but not ACE2, and that vitamin C remained unable to do so. Although prenol inhibited the entry of pseudotyped SARS-CoV-2, but not vesicular stomatitis virus, into human ACE2-expressing HEK293 cells, vitamin C blocked the entry of pseudotyped vesicular stomatitis virus, not SARS-CoV-2, indicating the specificity of the effect. Prenol, but not vitamin C, decreased SARS-CoV-2 spike S1-induced activation of NF-κB and the expression of proinflammatory cytokines in human A549 lung cells. Moreover, prenol also decreased the expression of proinflammatory cytokines induced by spike S1 of N501Y, E484K, Omicron, and Delta variants of SARS-CoV-2. Finally, oral treatment with prenol reduced fever, decreased lung inflammation, enhanced heart function, and improved locomotor activities in SARS-CoV-2 spike S1-intoxicated mice. These results suggest that prenol and prenol-containing fruits, but not vitamin C, may be more beneficial for fighting against COVID-19.
Collapse
Affiliation(s)
- Ramesh K. Paidi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Sumita Raha
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Rama K. Mishra
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, USA
| | - Brian Jeong
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Monica Sheinin
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA
| |
Collapse
|
17
|
Lee JW, Gu HO, Jung Y, Jung Y, Seo SY, Hong JH, Hong IS, Lee DH, Kim OH, Oh BC. Candesartan, an angiotensin-II receptor blocker, ameliorates insulin resistance and hepatosteatosis by reducing intracellular calcium overload and lipid accumulation. Exp Mol Med 2023:10.1038/s12276-023-00982-6. [PMID: 37121975 DOI: 10.1038/s12276-023-00982-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 01/17/2023] [Accepted: 01/30/2023] [Indexed: 05/02/2023] Open
Abstract
Insulin resistance is a major contributor to the pathogenesis of several human diseases, including type 2 diabetes, hypertension, and hyperlipidemia. Notably, insulin resistance and hypertension share common abnormalities, including increased oxidative stress, inflammation, and organelle dysfunction. Recently, we showed that excess intracellular Ca2+, a known pathogenic factor in hypertension, acts as a critical negative regulator of insulin signaling by forming Ca2+-phosphoinositides that prevent the membrane localization of AKT, a key serine/threonine kinase signaling molecule. Whether preventing intracellular Ca2+ overload improves insulin sensitivity, however, has not yet been investigated. Here, we show that the antihypertensive agent candesartan, compared with other angiotensin-II receptor blockers, has previously unrecognized beneficial effects on attenuating insulin resistance. We found that candesartan markedly reduced palmitic acid (PA)-induced intracellular Ca2+ overload and lipid accumulation by normalizing dysregulated store-operated channel (SOC)-mediated Ca2+ entry into cells, which alleviated PA-induced insulin resistance by promoting insulin-stimulated AKT membrane localization and increased the phosphorylation of AKT and its downstream substrates. As pharmacological approaches to attenuate intracellular Ca2+ overload in vivo, administering candesartan to obese mice successfully decreased insulin resistance, hepatic steatosis, dyslipidemia, and tissue inflammation by inhibiting dysregulated SOC-mediated Ca2+ entry and ectopic lipid accumulation. The resulting alterations in the phosphorylation of key signaling molecules consequently alleviate impaired insulin signaling by increasing the postprandial membrane localization and phosphorylation of AKT. Thus, our findings provide robust evidence for the pleiotropic contribution of intracellular Ca2+ overload in the pathogenesis of insulin resistance and suggest that there are viable approved drugs that can be repurposed for the treatment of insulin resistance and hypertension.
Collapse
Affiliation(s)
- Jin Wook Lee
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, Gachon College of Medicine, Incheon, 21999, Republic of Korea
- Department of Health Sciences and Technology (GAIHST), Gachon University, Incheon, 21999, Republic of Korea
| | - Hyun-Oh Gu
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, Gachon College of Medicine, Incheon, 21999, Republic of Korea
- Department of Health Sciences and Technology (GAIHST), Gachon University, Incheon, 21999, Republic of Korea
| | - Yunshin Jung
- Department of Health Sciences and Technology (GAIHST), Gachon University, Incheon, 21999, Republic of Korea
| | - YunJae Jung
- Department of Health Sciences and Technology (GAIHST), Gachon University, Incheon, 21999, Republic of Korea
- Department of Microbiology, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon, 21999, Republic of Korea
| | - Seung-Yong Seo
- College of Pharmacy, Gachon University, Incheon, 21936, Republic of Korea
| | - Jeong-Hee Hong
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, Gachon College of Medicine, Incheon, 21999, Republic of Korea
| | - In-Sun Hong
- Department of Molecular Medicine, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon, 21999, Republic of Korea
| | - Dae Ho Lee
- Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, 21565, Republic of Korea
| | - Ok-Hee Kim
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, Gachon College of Medicine, Incheon, 21999, Republic of Korea.
| | - Byung-Chul Oh
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, Gachon College of Medicine, Incheon, 21999, Republic of Korea.
- Department of Health Sciences and Technology (GAIHST), Gachon University, Incheon, 21999, Republic of Korea.
| |
Collapse
|
18
|
Application of Chiral Piperidine Scaffolds in Drug Design. PHARMACEUTICAL FRONTS 2023. [DOI: 10.1055/s-0043-1764218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023] Open
Abstract
Chiral piperidine scaffolds are prevalent as the common cores of a large number of active pharmaceuticals in medical chemistry. This review outlined the diversity of chiral piperidine scaffolds in recently approved drugs, and also covers the scaffolds in leads and drug candidates. The significance of chiral piperidine scaffolds in drug design is also discussed in this article. With the introduction of chiral piperidine scaffolds into small molecules, the exploration of drug-like molecules can be benefitted from the following aspect: (1) modulating the physicochemical properties; (2) enhancing the biological activities and selectivity; (3) improving pharmacokinetic properties; and (4) reducing the cardiac hERG toxicity. Given above, chiral piperidine-based discovery of small molecules will be a promising strategy to enrich our molecules' library to fight against diseases.
Collapse
|
19
|
Liu S, Kormos BL, Knafels JD, Sahasrabudhe PV, Rosado A, Sommese RF, Reyes AR, Ward J, Roth Flach RJ, Wang X, Buzon LM, Reese MR, Bhattacharya SK, Omoto K, Filipski KJ. Structural studies identify angiotensin II receptor blocker-like compounds as branched-chain ketoacid dehydrogenase kinase inhibitors. J Biol Chem 2023; 299:102959. [PMID: 36717078 PMCID: PMC9976451 DOI: 10.1016/j.jbc.2023.102959] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 01/19/2023] [Accepted: 01/25/2023] [Indexed: 01/30/2023] Open
Abstract
The mammalian mitochondrial branched-chain ketoacid dehydrogenase (BCKD) complex is a multienzyme complex involved in the catabolism of branched-chain amino acids. BCKD is regulated by the BCKD kinase, or BCKDK, which binds to the E2 subunit of BCKD, phosphorylates its E1 subunit, and inhibits enzymatic activity. Inhibition of the BCKD complex results in increased levels of branched-chain amino acids and branched-chain ketoacids, and this buildup has been associated with heart failure, type 2 diabetes mellitus, and nonalcoholic fatty liver disease. To find BCKDK inhibitors for potential treatment of these diseases, we performed both NMR and virtual fragment screening and identified tetrazole-bearing fragments that bind BCKDK at multiple sites. Through structure-based virtual screening expanding from these fragments, the angiotensin receptor blocker class antihypertension drugs and angiotensin receptor blocker-like compounds were discovered to be potent BCKDK inhibitors, suggesting potential new avenues for heart failure treatment combining BCKDK inhibition and antihypertension.
Collapse
Affiliation(s)
- Shenping Liu
- Medicine Design, Pfizer Inc, Groton, Connecticut, USA.
| | | | | | | | - Amy Rosado
- Medicine Design, Pfizer Inc, Groton, Connecticut, USA
| | | | - Allan R Reyes
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Jessica Ward
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts, USA
| | | | - Xiaochun Wang
- Medicine Design, Pfizer Inc, Groton, Connecticut, USA
| | | | | | | | - Kiyoyuki Omoto
- Medicine Design, Pfizer Inc, Cambridge, Massachusetts, USA
| | | |
Collapse
|
20
|
Shi Y, Chen Y, Deng L, Du K, Lu S, Chen T. Structural Understanding of Peptide-Bound G Protein-Coupled Receptors: Peptide-Target Interactions. J Med Chem 2023; 66:1083-1111. [PMID: 36625741 DOI: 10.1021/acs.jmedchem.2c01309] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The activation of G protein-coupled receptors (GPCRs) is triggered by ligand binding to their orthosteric sites, which induces ligand-specific conformational changes. Agonists and antagonists bound to GPCR orthosteric sites provide detailed information on ligand-binding modes. Among these, peptide ligands play an instrumental role in GPCR pharmacology and have attracted increased attention as therapeutic drugs. The recent breakthrough in GPCR structural biology has resulted in the remarkable availability of peptide-bound GPCR complexes. Despite the several structural similarities shared by these receptors, they exhibit distinct features in terms of peptide recognition and receptor activation. From this perspective, we have summarized the current status of peptide-bound GPCR structural complexes, largely focusing on the interactions between the receptor and its peptide ligand at the orthosteric site. In-depth structural investigations have yielded valuable insights into the molecular mechanisms underlying peptide recognition. This study would contribute to the discovery of GPCR peptide drugs with improved therapeutic effects.
Collapse
Affiliation(s)
- Yuxin Shi
- School of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing 312000, China.,Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Yi Chen
- Department of Ultrasound Interventional, Eastern Hepatobiliary Surgery Hospital, Navy Medical University, Shanghai 200433, China
| | - Liping Deng
- School of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing 312000, China
| | - Kui Du
- School of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing 312000, China
| | - Shaoyong Lu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China.,Institute of Energy Metabolism and Health, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.,College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, China
| | - Ting Chen
- Department of Cardiology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| |
Collapse
|
21
|
Santra D, Banerjee A, De SK, Thatoi H, Maiti S. Relation of ACE2 with co-morbidity factors in SARS-CoV-2 pathogenicity. COMPARATIVE CLINICAL PATHOLOGY 2023; 32:179-189. [PMID: 36687210 PMCID: PMC9843654 DOI: 10.1007/s00580-023-03434-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 01/01/2023] [Indexed: 01/18/2023]
Abstract
In the last 3 years of the pandemic situation, SARS-CoV-2 caused a significant number of deaths. Infection rates for symptomatic and asymptomatic patients are higher than that for death. Eventually, researchers explored that the major deaths are attributed to several comorbidity factors. The confounding factors and gender-associated infection/death rate are observed globally. This suggests that SARS-CoV-2 selects the human system recognizing the internal comorbid environment. This article explored the influences of hypertension, diabetes, cardiovascular, and renovascular disorders in COVID-19 severity and mortality. Brief mechanistic layouts have been presented here, indicating some of the comorbidity as the critical determinant in the COVID-19 pathogenesis and related mortality.
Collapse
Affiliation(s)
- Dipannita Santra
- Department of Biochemistry and Biotechnology, Cell and Molecular Therapeutics Laboratory, Oriental Institute of Science and Technology, Midnapore, India
| | - Amrita Banerjee
- Department of Biochemistry and Biotechnology, Cell and Molecular Therapeutics Laboratory, Oriental Institute of Science and Technology, Midnapore, India
| | - Subrata Kr. De
- grid.412834.80000 0000 9152 1805Department of Zoology, Vidyasagar University, Midnapore, 721102 India ,grid.411552.60000 0004 1766 4022Mahatma Gandhi University, East Midnapore, WB India
| | - Hrudayanath Thatoi
- grid.444567.00000 0004 1801 0450Department of Biotechnology, North Orissa University, Sriram Chandra Vihar, Takatpur, Baripada India
| | - Smarajit Maiti
- Department of Biochemistry and Biotechnology, Cell and Molecular Therapeutics Laboratory, Oriental Institute of Science and Technology, Midnapore, India ,Agricure Biotech Research Society, Epidemiology and Human Health Division, Midnapore, 721101 India
| |
Collapse
|
22
|
Georgiadis D, Skoulikas N, Papakyriakou A, Stratikos E. Phosphinic Peptides as Tool Compounds for the Study of Pharmacologically Relevant Zn-Metalloproteases. ACS Pharmacol Transl Sci 2022; 5:1228-1253. [PMID: 36524013 PMCID: PMC9745897 DOI: 10.1021/acsptsci.2c00183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Indexed: 11/29/2022]
Abstract
Phosphinic peptides constitute an important class of bioactive compounds that have found a wide range of applications in the field of biology and pharmacology of Zn-metalloproteases, the largest family of proteases in humans. They are designed to mimic the structure of natural substrates during their proteolysis, thus acting as mechanism-based, transition state analogue inhibitors. A combination of electrostatic interactions between the phosphinic acid group and the Zn cation as well as optimal noncovalent enzyme-ligand interactions can result in both high binding affinity for the desired target and selectivity against other proteases. Due to these unique properties, phosphinic peptides have been mainly employed as tool compounds for (a) the purposes of rational drug design by serving as ligands in X-ray crystal structures of target enzymes and allowing the identification of crucial interactions that govern optimal molecular recognition, and (b) the delineation of biological pathways where Zn-metalloproteases are key regulators. For the latter objective, inhibitors of the phosphinopeptidic type have been used either unmodified or after being transformed to probes of various types, thus expanding the arsenal of functional tools available to researchers. The aim of this review is to summarize all recent research achievements in which phosphinic peptides have played a central role as tool compounds in the understanding of the mechanism and biological functions of Zn-metalloproteases in both health and disease.
Collapse
Affiliation(s)
- Dimitris Georgiadis
- Department
of Chemistry, National and Kapodistrian
University of Athens, GR-15784 Athens, Greece
| | - Nikolaos Skoulikas
- Department
of Chemistry, National and Kapodistrian
University of Athens, GR-15784 Athens, Greece
| | - Athanasios Papakyriakou
- National
Centre for Scientific Research “Demokritos”, Agia Paraskevi GR-15341 Athens, Greece
| | - Efstratios Stratikos
- Department
of Chemistry, National and Kapodistrian
University of Athens, GR-15784 Athens, Greece
- National
Centre for Scientific Research “Demokritos”, Agia Paraskevi GR-15341 Athens, Greece
| |
Collapse
|
23
|
Ćwilichowska N, Świderska KW, Dobrzyń A, Drąg M, Poręba M. Diagnostic and therapeutic potential of protease inhibition. Mol Aspects Med 2022; 88:101144. [PMID: 36174281 DOI: 10.1016/j.mam.2022.101144] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 07/20/2022] [Accepted: 09/09/2022] [Indexed: 12/14/2022]
Abstract
Proteases are enzymes that hydrolyze peptide bonds in proteins and peptides; thus, they control virtually all biological processes. Our understanding of protease function has advanced considerably from nonselective digestive enzymes to highly specialized molecular scissors that orchestrate complex signaling networks through a limited proteolysis. The catalytic activity of proteases is tightly regulated at several levels, ranging from gene expression through trafficking and maturation to posttranslational modifications. However, when this delicate balance is disturbed, many diseases develop, including cancer, inflammatory disorders, diabetes, and neurodegenerative diseases. This new understanding of the role of proteases in pathologic physiology indicates that these enzymes represent excellent molecular targets for the development of therapeutic inhibitors, as well as for the design of chemical probes to visualize their redundant activity. Recently, numerous platform technologies have been developed to identify and optimize protease substrates and inhibitors, which were further used as lead structures for the development of chemical probes and therapeutic drugs. Due to this considerable success, the clinical potential of proteases in therapeutics and diagnostics is rapidly growing and is still not completely explored. Therefore, small molecules that can selectively target aberrant protease activity are emerging in diseases cells. In this review, we describe modern trends in the design of protease drugs as well as small molecule activity-based probes to visualize selected proteases in clinical settings.
Collapse
Affiliation(s)
- Natalia Ćwilichowska
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland
| | - Karolina W Świderska
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland
| | - Agnieszka Dobrzyń
- Nencki Institute of Experimental Biology, Ludwika Pasteura 3, 02-093, Warsaw, Poland
| | - Marcin Drąg
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland.
| | - Marcin Poręba
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland.
| |
Collapse
|
24
|
Sheinin M, Jeong B, Paidi RK, Pahan K. Regression of Lung Cancer in Mice by Intranasal Administration of SARS-CoV-2 Spike S1. Cancers (Basel) 2022; 14:5648. [PMID: 36428739 PMCID: PMC9688283 DOI: 10.3390/cancers14225648] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 11/18/2022] Open
Abstract
This study underlines the importance of SARS-CoV-2 spike S1 in prompting death in cultured non-small cell lung cancer (NSCLC) cells and in vivo in lung tumors in mice. Interestingly, we found that recombinant spike S1 treatment at very low doses led to death of human A549 NSCLC cells. On the other hand, boiled recombinant SARS-CoV-2 spike S1 remained unable to induce death, suggesting that the induction of cell death in A549 cells was due to native SARS-CoV-2 spike S1 protein. SARS-CoV-2 spike S1-induced A549 cell death was also inhibited by neutralizing antibodies against spike S1 and ACE2. Moreover, our newly designed wild type ACE2-interacting domain of SARS-CoV-2 (wtAIDS), but not mAIDS, peptide also attenuated SARS-CoV-2 spike S1-induced cell death, suggesting that SARS-CoV-2 spike S1-induced death in A549 NSCLC cells depends on its interaction with ACE2 receptor. Similarly, recombinant spike S1 treatment also led to death of human H1299 and H358 NSCLC cells. Finally, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) intoxication led to the formation tumors in lungs of A/J mice and alternate day intranasal treatment with low dose of recombinant SARS-CoV-2 spike S1 from 22-weeks of NNK insult (late stage) induced apoptosis and tumor regression in the lungs. These studies indicate that SARS-CoV-2 spike S1 may have implications for lung cancer treatment.
Collapse
Affiliation(s)
- Monica Sheinin
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Brian Jeong
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Ramesh K. Paidi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
25
|
Guimarães-Nobre CC, Mendonça-Reis E, Teixeira-Alves LR, Miranda-Alves L, Berto-Junior C. ATR1 Angiotensin II Receptor Reduces Hemoglobin S Polymerization, Phosphatidylserine Exposure, and Increases Deformability of Sickle Cell Disease Erythrocytes. Cell Biochem Biophys 2022; 80:711-721. [PMID: 36175813 DOI: 10.1007/s12013-022-01096-y] [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: 06/30/2022] [Accepted: 09/11/2022] [Indexed: 11/26/2022]
Abstract
Angiotensin II (Ang II) regulates blood volume and stimulates erythropoiesis through AT1 (ATR1) and AT2 (ATR2) receptors, found in multiple tissues, including erythrocytes. Sickle cell disease (SCD) patients present altered Ang II levels. Hemoglobin S polymerization, deformability and phosphatidylserine translocation are important features of mature erythrocytes, therefore, our hypothesis is Ang II affects these parameters and, if it does, what would be the influence of AT1R and AT2R on these effects. A polymerization assay (PA), deformability, and annexin V binding were performed in SCD erythrocytes samples adding Ang II, ATR1 antagonist (losartan or eprosartan), and ATR2 antagonist (PD123319). Through the PA test, we observed a dose-dependent polymerization inhibition effect when comparing Ang II to control. Losartan did not affect the level or the rate of Ang II inhibition, while PD123319 showed an increased level of protection against polymerization, and eprosartan brought levels back to control. Ang II was able to reduce the translocation of phosphatidylserine from the inner to the outer leaflet, a marker of eryptosis, in the presence of PD123319. Also, ATR1 showed a positive effect increasing deformability. Our data shows that ATR1 is important for maintenance of erythrocyte physiological function in SCD and for prolonging its life.
Collapse
Affiliation(s)
- Camila Cristina Guimarães-Nobre
- Grupo de Pesquisa em Fisiologia Eritróide - GPFisEri, Universidade Federal do Rio de Janeiro, Campus Macaé, Rio de Janeiro, Brazil
- Programa de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Evelyn Mendonça-Reis
- Grupo de Pesquisa em Fisiologia Eritróide - GPFisEri, Universidade Federal do Rio de Janeiro, Campus Macaé, Rio de Janeiro, Brazil
- Programa de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lyzes Rosa Teixeira-Alves
- Grupo de Pesquisa em Fisiologia Eritróide - GPFisEri, Universidade Federal do Rio de Janeiro, Campus Macaé, Rio de Janeiro, Brazil
- Programa de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leandro Miranda-Alves
- Programa de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Endocrinologia Experimental- LEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clemilson Berto-Junior
- Grupo de Pesquisa em Fisiologia Eritróide - GPFisEri, Universidade Federal do Rio de Janeiro, Campus Macaé, Rio de Janeiro, Brazil.
- Programa de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
- Laboratório de Endocrinologia Experimental- LEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
26
|
Chepilo DA, Gegechkori VI, Shchepochkina OY, Efremov AY, Levko AA, Chadova NN, Shestakov VN. A Complex Approach to the Determination of Authenticity in the Development of Standard Samples for Ace Inhibitors. Pharm Chem J 2022. [DOI: 10.1007/s11094-022-02667-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
Deng Z, Gao X, Utsunomiya H, Arner JW, Ruzbarsky JJ, Huard M, Ravuri S, Philippon MJ, Huard J. Effects of oral losartan administration on homeostasis of articular cartilage and bone in a rabbit model. Bone Rep 2022; 16:101526. [PMID: 35372645 PMCID: PMC8971351 DOI: 10.1016/j.bonr.2022.101526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 11/26/2022] Open
Abstract
Background and aims Previous work has shown that oral losartan can enhance microfracture-mediated cartilage repair in a rabbit osteochondral defect injury model. In this study, we aimed to determine whether oral losartan would have a detrimental effect on articular cartilage and bone homeostasis in the uninjured sides. Methods New Zealand rabbits were divided into 4 groups including normal uninjured (Normal), contralateral uninjured side of osteochondral defect (Defect), osteochondral defect plus microfracture (Microfracture) and osteochondral defect plus microfracture and losartan oral administration (10 mg/kg/day) (Losartan). Rabbits underwent different surgeries and treatment and were sacrificed at 12 weeks. Both side of the normal group and uninjured side of treatment groups tibias were harvested for Micro-CT and histological analysis for cartilage and bone including H&E staining, Herovici's staining (bone and cartilage) Alcian blue and Safranin O staining (cartilage) as well as immunohistochemistry of losartan related signaling pathways molecules for both cartilage and bone. Results Our results showed losartan oral treatment at 10 mg/kg/day slightly increase Alcian blue positive matrix as well as decrease collagen type 3 in articular cartilage while having no significant effect on articular cartilage structure, cellularity, and other matrix. Losartan treatment also did not affect angiotensin receptor type 1 (AGTR1), angiotensin receptor type 2 (AGTR2) and phosphorylated transforming factor β1 activated kinase 1 (pTAK1) expression level and pattern in the articular cartilage. Furthermore, losartan treatment did not affect microarchitecture of normal cancellous bone and cortical bone of tibias compared to normal and other groups. Losartan treatment slightly increased osteocalcin positive osteoblasts on the surface of cancellous bone and did not affect bone matrix collagen type 1 content and did not change AGTR1, AGTR2 and pTAK1 signal molecule expression. Conclusion Oral losartan used as a microfracture augmentation therapeutic does not have significant effect on uninjured articular cartilage and bone based on our preclinical rabbit model. These results provided further evidence that the current regimen of using losartan as a microfracture augmentation therapeutic is safe with respect to bone and cartilage homeostasis and support clinical trials for its application in human cartilage repair.
Collapse
|
28
|
Cong X, Zhang X, Liang X, He X, Tang Y, Zheng X, Lu S, Zhang J, Chen T. Delineating the conformational landscape and intrinsic properties of the angiotensin II type 2 receptor using a computational study. Comput Struct Biotechnol J 2022; 20:2268-2279. [PMID: 35615027 PMCID: PMC9117689 DOI: 10.1016/j.csbj.2022.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 12/22/2022] Open
Abstract
As a key regulator for the renin-angiotensin system, a class A G protein-coupled receptor (GPCR), AngII type 2 receptor (AT2R), plays a pivotal role in the homeostasis of the cardiovascular system. Compared with other GPCRs, AT2R has a unique antagonist-bound conformation and its mechanism is still an enigma. Here, we applied combined dynamic and evolutional approaches to investigate the conformational space and intrinsic properties of AT2R. With molecular dynamic simulations, Markov State Models, and statistics coupled analysis, we captured the conformational landscape of AT2R and identified its uniquity from both dynamical and evolutional viewpoints. A cryptic pocket was also discovered in the intermediate state during conformation transitions. These findings offer a deeper understanding of the AT2R mechanism at an atomic level and provide hints for the design of novel AT2R modulators.
Collapse
Affiliation(s)
- Xiaoliang Cong
- Department of Cardiology, Shanghai Changzheng Hospital, the Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Xiaogang Zhang
- Department of Cardiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Xin Liang
- Department of Cardiology, Shanghai Changzheng Hospital, the Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Xinheng He
- Medicinal Chemistry and Bioinformatics Centre, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Yehua Tang
- Department of Cardiology, Shanghai Changzheng Hospital, the Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Xing Zheng
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Shaoyong Lu
- Medicinal Chemistry and Bioinformatics Centre, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
- Corresponding authors.
| | - Jiayou Zhang
- Department of Cardiology, Shanghai Changzheng Hospital, the Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
- Corresponding authors.
| | - Ting Chen
- Department of Cardiology, Shanghai Changzheng Hospital, the Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
- Corresponding authors.
| |
Collapse
|
29
|
Alvarenga DJ, Matias LMF, Cordeiro CF, Souza TBD, Lavorato SN, Pereira MGAG, Dias DF, Carvalho DT. Synthesis of eugenol-derived glucosides and evaluation of their ability in inhibiting the angiotensin converting enzyme. Nat Prod Res 2022; 36:2246-2253. [PMID: 33032468 DOI: 10.1080/14786419.2020.1827399] [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] [Indexed: 10/23/2022]
Abstract
We report here a series of glucosides which are active as inhibitors of the angiotensin converting enzyme (ACE). They are structurally related to the natural compound eugenol and exhibited significant inhibition values. Their syntheses were expeditious and we could obtain informative docking plots of them complexed to this enzyme. A glucoside derived from eugenol, carrying a carboxylic group in the aglycone, was the most active of them (with an IC50 of 0.4 mM) and showed good binding energies in docking studies with ACE. Moreover, computational prediction of toxicity risks, physicochemical properties and drug score show that the glucoside derivative of eugenol is a suitable compound for optimisation studies aimed at finding new drug candidates.
Collapse
Affiliation(s)
| | | | | | | | - Stefânia Neiva Lavorato
- Centro das Ciências Biológicas e da Saúde, Universidade Federal do Oeste da Bahia, Barreiras, BA, Brazil
| | | | | | | |
Collapse
|
30
|
Kanno Y, Shu E. α2-Antiplasmin as a Potential Therapeutic Target for Systemic Sclerosis. Life (Basel) 2022; 12:life12030396. [PMID: 35330147 PMCID: PMC8953682 DOI: 10.3390/life12030396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 11/24/2022] Open
Abstract
Systemic sclerosis is a connective tissue disease of unknown origin that is characterized by immune system abnormalities, vascular damage, and extensive fibrosis of the skin and visceral organs. α2-antiplasmin is known to be the main plasmin inhibitor and has various functions such as cell differentiation and cytokine production, as well as the regulation of the maintenance of the immune system, endothelial homeostasis, and extracellular matrix metabolism. The expression of α2-antiplasmin is elevated in dermal fibroblasts from systemic sclerosis patients, and the blockade of α2-antiplasmin suppresses fibrosis progression and vascular dysfunction in systemic sclerosis model mice. α2-antiplasmin may have promise as a potential therapeutic target for systemic sclerosis. This review considers the role of α2-antiplasmin in the progression of systemic sclerosis.
Collapse
Affiliation(s)
- Yosuke Kanno
- Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women’s College of Liberal Arts, 97-1 Kodo Kyotanabe, Kyoto 610-0395, Japan
- Department of Dermatology, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan;
- Correspondence: ; Tel.:+81-0774-65-8629
| | - En Shu
- Department of Dermatology, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan;
| |
Collapse
|
31
|
Banik SK, Baishya S, Das Talukdar A, Choudhury MD. Network analysis of atherosclerotic genes elucidates druggable targets. BMC Med Genomics 2022; 15:42. [PMID: 35241081 PMCID: PMC8893053 DOI: 10.1186/s12920-022-01195-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/18/2021] [Indexed: 11/22/2022] Open
Abstract
Background Atherosclerosis is one of the major causes of cardiovascular disease. It is characterized by the accumulation of atherosclerotic plaque in arteries under the influence of inflammatory responses, proliferation of smooth muscle cell, accumulation of modified low density lipoprotein. The pathophysiology of atherosclerosis involves the interplay of a number of genes and metabolic pathways. In traditional translation method, only a limited number of genes and pathways can be studied at once. However, the new paradigm of network medicine can be explored to study the interaction of a large array of genes and their functional partners and their connections with the concerned disease pathogenesis. Thus, in our study we employed a branch of network medicine, gene network analysis as a tool to identify the most crucial genes and the miRNAs that regulate these genes at the post transcriptional level responsible for pathogenesis of atherosclerosis. Result From NCBI database 988 atherosclerotic genes were retrieved. The protein–protein interaction using STRING database resulted in 22,693 PPI interactions among 872 nodes (genes) at different confidence score. The cluster analysis of the 872 genes using MCODE, a plug-in of Cytoscape software revealed a total of 18 clusters, the topological parameter and gene ontology analysis facilitated in the selection of four influential genes viz., AGT, LPL, ITGB2, IRS1 from cluster 3. Further, the miRNAs (miR-26, miR-27, and miR-29 families) targeting these genes were obtained by employing MIENTURNET webtool. Conclusion Gene network analysis assisted in filtering out the 4 probable influential genes and 3 miRNA families in the pathogenesis of atherosclerosis. These genes, miRNAs can be targeted to restrict the occurrence of atherosclerosis. Given the importance of atherosclerosis, any approach in the understanding the genes involved in its pathogenesis can substantially enhance the health care system. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01195-y.
Collapse
Affiliation(s)
- Sheuli Kangsa Banik
- Department of Life Science and Bioinformatics, Assam University, Silchar, India
| | - Somorita Baishya
- Department of Life Science and Bioinformatics, Assam University, Silchar, India
| | - Anupam Das Talukdar
- Department of Life Science and Bioinformatics, Assam University, Silchar, India
| | | |
Collapse
|
32
|
Macroalgal Proteins: A Review. Foods 2022; 11:foods11040571. [PMID: 35206049 PMCID: PMC8871301 DOI: 10.3390/foods11040571] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/31/2022] [Accepted: 02/08/2022] [Indexed: 12/11/2022] Open
Abstract
Population growth is the driving change in the search for new, alternative sources of protein. Macroalgae (otherwise known as seaweeds) do not compete with other food sources for space and resources as they can be sustainably cultivated without the need for arable land. Macroalgae are significantly rich in protein and amino acid content compared to other plant-derived proteins. Herein, physical and chemical protein extraction methods as well as novel techniques including enzyme hydrolysis, microwave-assisted extraction and ultrasound sonication are discussed as strategies for protein extraction with this resource. The generation of high-value, economically important ingredients such as bioactive peptides is explored as well as the application of macroalgal proteins in human foods and animal feed. These bioactive peptides that have been shown to inhibit enzymes such as renin, angiotensin-I-converting enzyme (ACE-1), cyclooxygenases (COX), α-amylase and α-glucosidase associated with hypertensive, diabetic, and inflammation-related activities are explored. This paper discusses the significant uses of seaweeds, which range from utilising their anthelmintic and anti-methane properties in feed additives, to food techno-functional ingredients in the formulation of human foods such as ice creams, to utilising their health beneficial ingredients to reduce high blood pressure and prevent inflammation. This information was collated following a review of 206 publications on the use of seaweeds as foods and feeds and processing methods to extract seaweed proteins.
Collapse
|
33
|
Paidi RK, Jana M, Mishra RK, Dutta D, Pahan K. Selective Inhibition of the Interaction between SARS-CoV-2 Spike S1 and ACE2 by SPIDAR Peptide Induces Anti-Inflammatory Therapeutic Responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:2521-2533. [PMID: 34645689 PMCID: PMC8664124 DOI: 10.4049/jimmunol.2100144] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 09/16/2021] [Indexed: 01/11/2023]
Abstract
Many patients with coronavirus disease 2019 in intensive care units suffer from cytokine storm. Although anti-inflammatory therapies are available to treat the problem, very often, these treatments cause immunosuppression. Because angiotensin-converting enzyme 2 (ACE2) on host cells serves as the receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), to delineate a SARS-CoV-2-specific anti-inflammatory molecule, we designed a hexapeptide corresponding to the spike S1-interacting domain of ACE2 receptor (SPIDAR) that inhibited the expression of proinflammatory molecules in human A549 lung cells induced by pseudotyped SARS-CoV-2, but not vesicular stomatitis virus. Accordingly, wild-type (wt), but not mutated (m), SPIDAR inhibited SARS-CoV-2 spike S1-induced activation of NF-κB and expression of IL-6 and IL-1β in human lung cells. However, wtSPIDAR remained unable to reduce activation of NF-κB and expression of proinflammatory molecules in lungs cells induced by TNF-α, HIV-1 Tat, and viral dsRNA mimic polyinosinic-polycytidylic acid, indicating the specificity of the effect. The wtSPIDAR, but not mutated SPIDAR, also hindered the association between ACE2 and spike S1 of SARS-CoV-2 and inhibited the entry of pseudotyped SARS-CoV-2, but not vesicular stomatitis virus, into human ACE2-expressing human embryonic kidney 293 cells. Moreover, intranasal treatment with wtSPIDAR, but not mutated SPIDAR, inhibited lung activation of NF-κB, protected lungs, reduced fever, improved heart function, and enhanced locomotor activities in SARS-CoV-2 spike S1-intoxicated mice. Therefore, selective targeting of SARS-CoV-2 spike S1-to-ACE2 interaction by wtSPIDAR may be beneficial for coronavirus disease 2019.
Collapse
Affiliation(s)
- Ramesh K Paidi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL
| | - Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL
| | - Rama K Mishra
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL; and
| | - Debashis Dutta
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL;
- Division of Research and Development, Jesse Brown VA Medical Center, Chicago, IL
| |
Collapse
|
34
|
Paidi RK, Jana M, Raha S, McKay M, Sheinin M, Mishra RK, Pahan K. Eugenol, a Component of Holy Basil (Tulsi) and Common Spice Clove, Inhibits the Interaction Between SARS-CoV-2 Spike S1 and ACE2 to Induce Therapeutic Responses. J Neuroimmune Pharmacol 2021; 16:743-755. [PMID: 34677731 PMCID: PMC8531902 DOI: 10.1007/s11481-021-10028-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022]
Abstract
Spike S1 of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) binds to angiotensin-converting enzyme 2 (ACE2) on host cells to enter the cell and initiate COVID-19. Since ACE2 is a favorable enzyme, we were interested in finding a molecule capable of binding spike S1, but not ACE2, and inhibiting the interaction between spike S1 and ACE2. Holy basil (Tulsi) has a long history as a medicine for different human disorders. Therefore, we screened different components of Tulsi leaf and found that eugenol, but not other major components (e.g. ursolic acid, oleanolic acid and β-caryophylline), inhibited the interaction between spike S1 and ACE2 in an AlphaScreen-based assay. By in silico analysis and thermal shift assay, we also observed that eugenol associated with spike S1, but not ACE2. Accordingly, eugenol strongly suppressed the entry of pseudotyped SARS-CoV-2, but not vesicular stomatitis virus (VSV), into human ACE2-expressing HEK293 cells. Eugenol also reduced SARS-CoV-2 spike S1-induced activation of NF-κB and the expression of IL-6, IL-1β and TNFα in human A549 lung cells. Moreover, oral treatment with eugenol reduced lung inflammation, decreased fever, improved heart function, and enhanced locomotor activities in SARS-CoV-2 spike S1-intoxicated mice. Therefore, selective targeting of SARS-CoV-2 spike S1, but not ACE2, by eugenol may be beneficial for COVID-19 treatment.
Collapse
Affiliation(s)
- Ramesh Kumar Paidi
- Department of Neurological Sciences, Rush University Medical Center, IL, Chicago, USA
| | - Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, IL, Chicago, USA
| | - Sumita Raha
- Department of Neurological Sciences, Rush University Medical Center, IL, Chicago, USA
| | - Mary McKay
- Department of Neurological Sciences, Rush University Medical Center, IL, Chicago, USA
| | - Monica Sheinin
- Department of Neurological Sciences, Rush University Medical Center, IL, Chicago, USA
| | - Rama K Mishra
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, IL, Chicago, USA. .,Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA.
| |
Collapse
|
35
|
Pucci F, Annoni F, dos Santos RAS, Taccone FS, Rooman M. Quantifying Renin-Angiotensin-System Alterations in COVID-19. Cells 2021; 10:2755. [PMID: 34685735 PMCID: PMC8535134 DOI: 10.3390/cells10102755] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/17/2022] Open
Abstract
The renin-angiotensin system (RAS) plays a pivotal role in a wide series of physiological processes, among which inflammation and blood pressure regulation. One of its key components, the angiotensin-converting enzyme 2, has been identified as the entry point of the SARS-CoV-2 virus into the host cells, and therefore a lot of research has been devoted to study RAS dysregulation in COVID-19. Here we discuss the alterations of the regulatory RAS axes due to SARS-CoV-2 infection on the basis of a series of recent clinical investigations and experimental analyzes quantifying, e.g., the levels and activity of RAS components. We performed a comprehensive meta-analysis of these data in view of disentangling the links between the impaired RAS functioning and the pathophysiological characteristics of COVID-19. We also review the effects of several RAS-targeting drugs and how they could potentially help restore the normal RAS functionality and minimize the COVID-19 severity. Finally, we discuss the conflicting evidence found in the literature and the open questions on RAS dysregulation in SARS-CoV-2 infection whose resolution would improve our understanding of COVID-19.
Collapse
Affiliation(s)
- Fabrizio Pucci
- 3BIO—Computational Biology and Bioinformatics, Université Libre de Bruxelles, 1050 Brussels, Belgium;
- (IB)—Interuniversity Institute of Bioinformatics in Brussels, 1050 Brussels, Belgium
| | - Filippo Annoni
- Department of Intensive Care, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium; (F.A.); (F.S.T.)
| | | | - Fabio Silvio Taccone
- Department of Intensive Care, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium; (F.A.); (F.S.T.)
| | - Marianne Rooman
- 3BIO—Computational Biology and Bioinformatics, Université Libre de Bruxelles, 1050 Brussels, Belgium;
- (IB)—Interuniversity Institute of Bioinformatics in Brussels, 1050 Brussels, Belgium
| |
Collapse
|
36
|
Chen CY, Tsai TY, Chen BH. Effects of Black Garlic Extract and Nanoemulsion on the Deoxy Corticosterone Acetate-Salt Induced Hypertension and Its Associated Mild Cognitive Impairment in Rats. Antioxidants (Basel) 2021; 10:1611. [PMID: 34679745 PMCID: PMC8533483 DOI: 10.3390/antiox10101611] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 11/16/2022] Open
Abstract
Organosulfur compounds, phenolic acids and flavonoids in raw and black garlic were determined, and followed by preparation of black garlic nanoemulsion for studying their effects on deoxycorticosterone acetate-salt-induced hypertension and associated mild cognitive impairment in rats. Three organosulfur compounds, including diallyl sulfide (87.8 μg/g), diallyl disulfide (203.9 μg/g) and diallyl trisulfide (282.6 μg/g) were detected in black garlic by GC-MS, while gallic acid (19.19 μg/g), p-coumaric acid (27.03 μg/g) and quercetin (22.77 μg/g) were detected by UPLC-MS/MS. High doses of both black garlic extract and nanoemulsion prepared using Tween-80, glycerol, grapeseed oil and water could decrease systolic blood pressure through the elevation of bradykinin and nitric oxide levels as well as diminish aldosterone and angiotensin II levels in rats. In Morris water maze test, they could significantly decrease escape latency and swimming distance and increase the time spent in the target quadrant, accompanied by a decline of acetylcholinesterase activity and malondialdehyde level in the hippocampus as well as a rise in glutathione level and activities of superoxide dismutase, catalase and glutathione peroxidase. In addition, the levels of tumor necrosis factor, interleukin-6 and interleukin-1β were reduced. Effects of lowering blood pressure and improving learning/memory ability in rats followed the order: lisinopril > black garlic nanoemulsion > black garlic extract.
Collapse
Affiliation(s)
- Chun-Yu Chen
- Department of Food Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan; (C.-Y.C.); (T.-Y.T.)
| | - Tsung-Yu Tsai
- Department of Food Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan; (C.-Y.C.); (T.-Y.T.)
| | - Bing-Huei Chen
- Department of Food Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan; (C.-Y.C.); (T.-Y.T.)
- Department of Nutrition, China Medical University, Taichung 40401, Taiwan
| |
Collapse
|
37
|
Barbiero P, Viñas Torné R, Lió P. Graph Representation Forecasting of Patient's Medical Conditions: Toward a Digital Twin. Front Genet 2021; 12:652907. [PMID: 34603366 PMCID: PMC8481902 DOI: 10.3389/fgene.2021.652907] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/24/2021] [Indexed: 01/05/2023] Open
Abstract
Objective: Modern medicine needs to shift from a wait and react, curative discipline to a preventative, interdisciplinary science aiming at providing personalized, systemic, and precise treatment plans to patients. To this purpose, we propose a "digital twin" of patients modeling the human body as a whole and providing a panoramic view over individuals' conditions. Methods: We propose a general framework that composes advanced artificial intelligence (AI) approaches and integrates mathematical modeling in order to provide a panoramic view over current and future pathophysiological conditions. Our modular architecture is based on a graph neural network (GNN) forecasting clinically relevant endpoints (such as blood pressure) and a generative adversarial network (GAN) providing a proof of concept of transcriptomic integrability. Results: We tested our digital twin model on two simulated clinical case studies combining information at organ, tissue, and cellular level. We provided a panoramic overview over current and future patient's conditions by monitoring and forecasting clinically relevant endpoints representing the evolution of patient's vital parameters using the GNN model. We showed how to use the GAN to generate multi-tissue expression data for blood and lung to find associations between cytokines conditioned on the expression of genes in the renin-angiotensin pathway. Our approach was to detect inflammatory cytokines, which are known to have effects on blood pressure and have previously been associated with SARS-CoV-2 infection (e.g., CXCR6, XCL1, and others). Significance: The graph representation of a computational patient has potential to solve important technological challenges in integrating multiscale computational modeling with AI. We believe that this work represents a step forward toward next-generation devices for precision and predictive medicine.
Collapse
|
38
|
Qi Z, Wang T, Chen X, Wong CK, Ding Q, Sauer H, Chen ZF, Long C, Yao X, Cai Z, Tsang SY. Extracellular and Intracellular Angiotensin II Regulate the Automaticity of Developing Cardiomyocytes via Different Signaling Pathways. Front Mol Biosci 2021; 8:699827. [PMID: 34513920 PMCID: PMC8425478 DOI: 10.3389/fmolb.2021.699827] [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: 04/24/2021] [Accepted: 07/27/2021] [Indexed: 12/02/2022] Open
Abstract
Angiotensin II (Ang II) plays an important role in regulating various physiological processes. However, little is known about the existence of intracellular Ang II (iAng II), whether iAng II would regulate the automaticity of early differentiating cardiomyocytes, and the underlying mechanism involved. Here, iAng II was detected by immunocytochemistry and ultra-high performance liquid chromatography combined with electrospray ionization triple quadrupole tandem mass spectrometry in mouse embryonic stem cell–derived cardiomyocytes (mESC-CMs) and neonatal rat ventricular myocytes. Expression of AT1R-YFP in mESC-CMs revealed that Ang II type 1 receptors were located on the surface membrane, while immunostaining of Ang II type 2 receptors (AT2R) revealed that AT2R were predominately located on the nucleus and the sarcoplasmic reticulum. While extracellular Ang II increased spontaneous action potentials (APs), dual patch clamping revealed that intracellular delivery of Ang II or AT2R activator C21 decreased spontaneous APs. Interestingly, iAng II was found to decrease the caffeine-induced increase in spontaneous APs and caffeine-induced calcium release, suggesting that iAng II decreased spontaneous APs via the AT2R- and ryanodine receptor–mediated pathways. This is the first study that provides evidence of the presence and function of iAng II in regulating the automaticity behavior of ESC-CMs and may therefore shed light on the role of iAng II in fate determination.
Collapse
Affiliation(s)
- Zenghua Qi
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China.,Institute of Environmental Health and Pollution Control, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou, China
| | - Tao Wang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong, SAR China
| | - Xiangmao Chen
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Chun Kit Wong
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China
| | - Qianqian Ding
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China
| | - Heinrich Sauer
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Zhi-Feng Chen
- Institute of Environmental Health and Pollution Control, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou, China
| | - Cheng Long
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xiaoqiang Yao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong, SAR China
| | - Suk Ying Tsang
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China.,Key Laboratory for Regenerative Medicine, Ministry of Education, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China.,State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China
| |
Collapse
|
39
|
Ryu B, Kim YS, Jeon YJ. Seaweeds and Their Natural Products for Preventing Cardiovascular Associated Dysfunction. Mar Drugs 2021; 19:md19090507. [PMID: 34564168 PMCID: PMC8470597 DOI: 10.3390/md19090507] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/25/2022] Open
Abstract
Cardiovascular disease (CVD), which involves the onset and exacerbation of various conditions including dyslipidemia, activation of the renin-angiotensin system, vascular endothelial cell damage, and oxidative stress, is a leading cause of high mortality rates and accounts for one-third of deaths worldwide. Accordingly, as dietary changes in daily life are thought to greatly reduce the prevalence of CVD, numerous studies have been conducted to examine the potential use of foods and their bioactive components for preventing and treating CVD. In particular, seaweeds contain unique bioactive metabolites that are not found in terrestrial plants because of the harsh environment in which they survive, leading to in vitro and in vivo studies of their prevention and treatment effects. This review summarizes studies that focused on the beneficial effects of seaweeds and their natural products targeting markers involved in a cascade of mechanisms related to CVD pathogenesis. The purpose of this review is to describe the potential of seaweeds and their natural products for preventing and treating CVD based on in vivo and in vitro studies. This review provides a basis for future research in the field of marine drugs.
Collapse
Affiliation(s)
- Bomi Ryu
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea
- Correspondence: (B.R.); (Y.-J.J.); Tel.: +82-64-754-3475 (B.R. & Y.-J.J.)
| | - Young-Sang Kim
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea
- Marine Science Institute, Jeju National University, Jeju 63333, Korea
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea
- Marine Science Institute, Jeju National University, Jeju 63333, Korea
- Healthy Seafood Research Center, Jeju National University, Jeju 63243, Korea
- Correspondence: (B.R.); (Y.-J.J.); Tel.: +82-64-754-3475 (B.R. & Y.-J.J.)
| |
Collapse
|
40
|
Vikøren LA, Drotningsvik A, Midttun Ø, McCann A, Bergseth MT, Austgulen MH, Mellgren G, Ueland PM, Gudbrandsen OA. Baked cod consumption delayed the development of kidney and liver dysfunction and affected plasma amino acid concentrations, but did not affect blood pressure, blood glucose or liver triacylglycerol concentrations in obese fa/fa Zucker rats. Nutr Res 2021; 92:72-83. [PMID: 34274556 DOI: 10.1016/j.nutres.2021.05.009] [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: 11/26/2020] [Revised: 05/13/2021] [Accepted: 05/23/2021] [Indexed: 10/21/2022]
Abstract
Obesity is associated with changes in amino acid metabolism, and studies show that ingestion of fish proteins influence amino acid composition in plasma and urine, in addition to affecting risk factors for metabolic syndrome. Since the majority of fish proteins consumed by humans are as fish fillet, it is of interest to investigate if cod fillet intake affects amino acid composition and metabolic disorders. We hypothesized that a modified AIN-93G diet containing cod fillet would affect amino acid compositions in plasma and urine in obese rats, and also affect risk factors for metabolic syndrome when compared to rats fed a regular AIN-93G diet with casein as the protein source. Obese Zucker fa/fa rats, a rat model of metabolic syndrome, received diets containing 25% protein from lyophilized baked cod fillet and 75% protein from casein (Baked cod diet), or a Control diet with casein for four weeks. The Baked cod diet affected the amino acid composition in plasma, with e.g., lower glycine, histidine, homoarginine, homocysteine, methionine, proline and tyrosine concentrations, but did not affect amino acid concentrations in urine. The concentrations of markers for kidney and liver dysfunction were lower in the Baked cod group, however blood pressure development, fasting and postprandial glucose, and hepatic triacylglycerol concentrations were similar to the Control group. To conclude, substituting 25% of dietary protein with baked cod fillet affected concentrations of some amino acids in plasma and delayed development of kidney and liver dysfunction, but did not affect blood pressure, glucose concentration or fatty liver.
Collapse
Affiliation(s)
- Linn A Vikøren
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway; Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Aslaug Drotningsvik
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway
| | | | | | - Marthe T Bergseth
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway
| | - Maren H Austgulen
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Gunnar Mellgren
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Haukeland University Hospital, 5020 Bergen, Norway; Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, 5021 Bergen, Norway
| | | | - Oddrun A Gudbrandsen
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway.
| |
Collapse
|
41
|
Binder C, Poglitsch M, Duca F, Rettl R, Dachs TM, Dalos D, Schrutka L, Seirer B, Ligios LC, Capelle C, Eslam RB, Qin H, Hengstenberg C, Bonderman D. Renin Feedback Is an Independent Predictor of Outcome in HFpEF. J Pers Med 2021; 11:jpm11050370. [PMID: 34063595 PMCID: PMC8147649 DOI: 10.3390/jpm11050370] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/25/2021] [Accepted: 04/28/2021] [Indexed: 12/18/2022] Open
Abstract
Drugs which interact with the renin angiotensin aldosterone system (RAAS) aim to reduce the negative effects of angiotensin (Ang) II. Treatment with these drugs anticipate a compensatory up-regulation of renin; however, it has been shown that there is a large variability in circulating plasma renin (PRA), even in patients with optimal medical therapy in patients with heart failure (HF) with reduced ejection fraction (HFrEF). Our aim was to measure plasma renin activity (PRA-S), its response to RAAS inhibitor (RAASi) therapies and its effects on outcome in patients with HF with preserved ejection fraction (HFpEF). For this purpose, 150 HFpEF patients were included into a prospective single-center registry. Equilibrium (eq) angiotensin metabolites were measured from serum samples using mass spectroscopy. PRA-S (eqAng I + eqAng II) was calculated and compared in respect to the primary endpoint defined as all-cause death. PRA-S in patients with RAASi therapy was not significantly higher than in patients without RAASi (p = 0.262). Even after adjusting for confounding factors, PRA-S remained predictive for all-cause death in the multivariable model with a hazard ratio of 2.14 (95%CI 1.20–3.82, p = 0.010). We conclude that high PRA-S is associated with poor prognosis in patients with HFpEF, regardless of RAASi treatment, which could ultimately result in hyperactivated RAAS and consecutive negative effects on the cardiovascular and renal system, leading to poor outcome in patients with HFpEF.
Collapse
Affiliation(s)
- Christina Binder
- Division of Cardiology, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (F.D.); (R.R.); (T.M.D.); (D.D.); (L.S.); (B.S.); (L.C.L.); (C.C.); (R.B.E.); (H.Q.); (C.H.)
| | | | - Franz Duca
- Division of Cardiology, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (F.D.); (R.R.); (T.M.D.); (D.D.); (L.S.); (B.S.); (L.C.L.); (C.C.); (R.B.E.); (H.Q.); (C.H.)
| | - René Rettl
- Division of Cardiology, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (F.D.); (R.R.); (T.M.D.); (D.D.); (L.S.); (B.S.); (L.C.L.); (C.C.); (R.B.E.); (H.Q.); (C.H.)
| | - Theresa Marie Dachs
- Division of Cardiology, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (F.D.); (R.R.); (T.M.D.); (D.D.); (L.S.); (B.S.); (L.C.L.); (C.C.); (R.B.E.); (H.Q.); (C.H.)
| | - Daniel Dalos
- Division of Cardiology, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (F.D.); (R.R.); (T.M.D.); (D.D.); (L.S.); (B.S.); (L.C.L.); (C.C.); (R.B.E.); (H.Q.); (C.H.)
| | - Lore Schrutka
- Division of Cardiology, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (F.D.); (R.R.); (T.M.D.); (D.D.); (L.S.); (B.S.); (L.C.L.); (C.C.); (R.B.E.); (H.Q.); (C.H.)
| | - Benjamin Seirer
- Division of Cardiology, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (F.D.); (R.R.); (T.M.D.); (D.D.); (L.S.); (B.S.); (L.C.L.); (C.C.); (R.B.E.); (H.Q.); (C.H.)
| | - Luciana Camuz Ligios
- Division of Cardiology, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (F.D.); (R.R.); (T.M.D.); (D.D.); (L.S.); (B.S.); (L.C.L.); (C.C.); (R.B.E.); (H.Q.); (C.H.)
| | - Christophe Capelle
- Division of Cardiology, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (F.D.); (R.R.); (T.M.D.); (D.D.); (L.S.); (B.S.); (L.C.L.); (C.C.); (R.B.E.); (H.Q.); (C.H.)
| | - Roza Badr Eslam
- Division of Cardiology, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (F.D.); (R.R.); (T.M.D.); (D.D.); (L.S.); (B.S.); (L.C.L.); (C.C.); (R.B.E.); (H.Q.); (C.H.)
| | - Hong Qin
- Division of Cardiology, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (F.D.); (R.R.); (T.M.D.); (D.D.); (L.S.); (B.S.); (L.C.L.); (C.C.); (R.B.E.); (H.Q.); (C.H.)
| | - Christian Hengstenberg
- Division of Cardiology, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (F.D.); (R.R.); (T.M.D.); (D.D.); (L.S.); (B.S.); (L.C.L.); (C.C.); (R.B.E.); (H.Q.); (C.H.)
| | - Diana Bonderman
- Division of Cardiology, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (F.D.); (R.R.); (T.M.D.); (D.D.); (L.S.); (B.S.); (L.C.L.); (C.C.); (R.B.E.); (H.Q.); (C.H.)
- Correspondence: ; Tel.: +43-140-400-46-140
| |
Collapse
|
42
|
Dean AQ, Bozza WP, Twomey JD, Luo S, Nalli A, Zhang B. The fight against COVID-19: Striking a balance in the renin-angiotensin system. Drug Discov Today 2021; 26:2214-2220. [PMID: 33865979 PMCID: PMC8049177 DOI: 10.1016/j.drudis.2021.04.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 03/16/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022]
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) enters host cells by interacting with membrane-bound angiotensin-converting enzyme 2 (ACE2), a vital element in the renin–angiotensin system (RAS), which regulates blood pressure, fluid balance, and cardiovascular functions. We herein evaluate existing evidence for the molecular alterations within the RAS pathway (e.g., ACE2 and angiotensin II) during SARS-CoV-2 infection and subsequent Coronavirus Disease 2019 (COVID-19). This includes reports regarding potential effect of RAS blockade (e.g., ACE inhibitors and angiotensin II receptor blockers) on ACE2 expression and clinical outcomes in patients with co-morbidities commonly treated with these agents. The collective evidence suggests a dual role for ACE2 in COVID-19, depending on the stage of infection and the coexisting diseases in individual patients. This information is further discussed with respect to potential therapeutic strategies targeting RAS for COVID-19 treatment.
Collapse
Affiliation(s)
- Alexis Q Dean
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - William P Bozza
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Julianne D Twomey
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Shen Luo
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Ancy Nalli
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Baolin Zhang
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA.
| |
Collapse
|
43
|
Snake Venom Components: Tools and Cures to Target Cardiovascular Diseases. Molecules 2021; 26:molecules26082223. [PMID: 33921462 PMCID: PMC8070158 DOI: 10.3390/molecules26082223] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular diseases (CVDs) are considered as a major cause of death worldwide. Therefore, identifying and developing therapeutic strategies to treat and reduce the prevalence of CVDs is a major medical challenge. Several drugs used for the treatment of CVDs, such as captopril, emerged from natural products, namely snake venoms. These venoms are complex mixtures of bioactive molecules, which, among other physiological networks, target the cardiovascular system, leading to them being considered in the development and design of new drugs. In this review, we describe some snake venom molecules targeting the cardiovascular system such as phospholipase A2 (PLA2), natriuretic peptides (NPs), bradykinin-potentiating peptides (BPPs), cysteine-rich secretory proteins (CRISPs), disintegrins, fibrinolytic enzymes, and three-finger toxins (3FTXs). In addition, their molecular targets, and mechanisms of action—vasorelaxation, inhibition of platelet aggregation, cardioprotective activities—are discussed. The dissection of their biological effects at the molecular scale give insights for the development of future snake venom-derived drugs.
Collapse
|
44
|
De Pieri A, Korman BD, Jüngel A, Wuertz-Kozak K. Engineering Advanced In Vitro Models of Systemic Sclerosis for Drug Discovery and Development. Adv Biol (Weinh) 2021; 5:e2000168. [PMID: 33852183 PMCID: PMC8717409 DOI: 10.1002/adbi.202000168] [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: 06/30/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 12/19/2022]
Abstract
Systemic sclerosis (SSc) is a complex multisystem disease with the highest case-specific mortality among all autoimmune rheumatic diseases, yet without any available curative therapy. Therefore, the development of novel therapeutic antifibrotic strategies that effectively decrease skin and organ fibrosis is needed. Existing animal models are cost-intensive, laborious and do not recapitulate the full spectrum of the disease and thus commonly fail to predict human efficacy. Advanced in vitro models, which closely mimic critical aspects of the pathology, have emerged as valuable platforms to investigate novel pharmaceutical therapies for the treatment of SSc. This review focuses on recent advancements in the development of SSc in vitro models, sheds light onto biological (e.g., growth factors, cytokines, coculture systems), biochemical (e.g., hypoxia, reactive oxygen species) and biophysical (e.g., stiffness, topography, dimensionality) cues that have been utilized for the in vitro recapitulation of the SSc microenvironment, and highlights future perspectives for effective drug discovery and validation.
Collapse
Affiliation(s)
- Andrea De Pieri
- Dr. A. De Pieri, Prof. K. Wuertz-Kozak, Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY, 14623, USA
| | - Benjamin D Korman
- Prof. B. D. Korman, Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Astrid Jüngel
- Prof. A. Jüngel, Center of Experimental Rheumatology, University Clinic of Rheumatology, Balgrist University Hospital, University Hospital Zurich, Zurich, 8008, Switzerland
- Prof. A. Jüngel, Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Zurich, 8008, Switzerland
| | - Karin Wuertz-Kozak
- Dr. A. De Pieri, Prof. K. Wuertz-Kozak, Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY, 14623, USA
- Prof. K. Wuertz-Kozak, Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (Austria), Munich, 81547, Germany
| |
Collapse
|
45
|
Li HL, Yang SQ, Li XM, Li X, Wang BG. Structurally diverse alkaloids produced by Aspergillus creber EN-602, an endophytic fungus obtained from the marine red alga Rhodomela confervoides. Bioorg Chem 2021; 110:104822. [PMID: 33770672 DOI: 10.1016/j.bioorg.2021.104822] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 12/31/2022]
Abstract
Thirteen alkaloids, which include three new diketopiperazines, namely, 3-hydroxyprotuboxepin K (4), 3,15-dehydroprotuboxepin K (5), and versiamide A (6), together with ten known alkaloid derivatives (1-3 and 7-13), were isolated from the marine red algal-derived fungus Aspergillus creber EN-602. Versiamide A (6) represents the first example of a naturally occurring quinazolinone alkaloid with a diketopiperazine ring that is derived from phenylalanine (Phe) and leucine (Leu). The structures of these compounds were elucidated by detailed interpretation of their 1D/2D NMR spectroscopic and mass spectrometric data, while the absolute configurations of compounds 1-6 were established on the basis of X-ray crystallographic analysis and time-dependent density functional (TDDFT) calculations of the ECD spectra. Compounds 1, 2, and 4 exhibited inhibitory activity against the angiotensin converting enzyme (ACE) with IC50 values of 11.2, 16.0, and 22.4 μM, respectively, and compounds 5 and 6 inhibited various aquatic bacteria with MIC values that ranged from 8 to 64 μg/mL. The intermolecular interactions and potential binding sites between compounds 1-6 and ACE were investigated via molecular docking simulations.
Collapse
Affiliation(s)
- Hong-Lei Li
- CAS and Shandong Province Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Nanhai Road 7, Qingdao 266071, People's Republic of China; Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Wenhai Road 1, Qingdao 266237, People's Republic of China; Center for Ocean Mega-Science, Chinese Academy of Sciences, Nanhai Road 7, Qingdao 266071, People's Republic of China.
| | - Sui-Qun Yang
- CAS and Shandong Province Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Nanhai Road 7, Qingdao 266071, People's Republic of China; Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Wenhai Road 1, Qingdao 266237, People's Republic of China; Center for Ocean Mega-Science, Chinese Academy of Sciences, Nanhai Road 7, Qingdao 266071, People's Republic of China
| | - Xiao-Ming Li
- CAS and Shandong Province Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Nanhai Road 7, Qingdao 266071, People's Republic of China; Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Wenhai Road 1, Qingdao 266237, People's Republic of China; Center for Ocean Mega-Science, Chinese Academy of Sciences, Nanhai Road 7, Qingdao 266071, People's Republic of China
| | - Xin Li
- CAS and Shandong Province Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Nanhai Road 7, Qingdao 266071, People's Republic of China; Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Wenhai Road 1, Qingdao 266237, People's Republic of China; Center for Ocean Mega-Science, Chinese Academy of Sciences, Nanhai Road 7, Qingdao 266071, People's Republic of China
| | - Bin-Gui Wang
- CAS and Shandong Province Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Nanhai Road 7, Qingdao 266071, People's Republic of China; Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Wenhai Road 1, Qingdao 266237, People's Republic of China; Center for Ocean Mega-Science, Chinese Academy of Sciences, Nanhai Road 7, Qingdao 266071, People's Republic of China.
| |
Collapse
|
46
|
Lite C, Ahmed SSSJ, Juliet M, Freddy AJ. SARS-CoV-2/human interactome reveals ACE2 locus crosstalk with the immune regulatory network in the host. Pathog Dis 2021; 79:6104547. [PMID: 33469663 PMCID: PMC7928900 DOI: 10.1093/femspd/ftab005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 01/15/2021] [Indexed: 01/28/2023] Open
Abstract
Severe acute respiratory syndrome, coronavirus 2 (SARS-CoV-2), remains to be a threat across the globe. SARS-CoV-2 entry into the host is mediated by binding of viral spike protein to the Human angiotensin-converting enzyme 2 (ACE2) receptor. ACE2 is an essential member of the Renin–Angiotensin system (RAS) involved in maintaining the blood pressure and vascular remodelling. Although ACE2 receptor is the entry point to the host, recent studies show activation of ACE2 to modulate the host to develop a suitable environment for its replication. However, the ACE2 activating the immune signals on SARS-CoV-2 attachment is still under investigation. We have used systems biological approach to construct the host regulatory network upon SARS-CoV-2 attachment to the ACE2 receptor. Since lungs are the primary infection site, we integrate human lung gene expression profile along with the host regulatory network to demonstrate the altered host signalling mechanism in viral infection. Further, the network was functionally enriched to determine immune modulation in the network. We also used the proteomic database to assess the occurrence of similar signalling events in other human tissues that exhibit lineage of infection across different organs. The constructed network contains 133 host proteins with 298 interactions that directly or indirectly connect to the ACE2 receptor. Among 133 proteins, 29 were found to be differentially regulated in the host lungs on SARS-CoV-2 infection. Altered proteins connect multiple proteins in a network that modulates kinase, carboxypeptidase and cytokine activity, leading to changes in the host immune system, cell cycle and signal transduction mechanisms. Further investigation showed the presence of similar signalling events in the kidneys, placenta, pancreas, testis, small intestine and adrenal gland as well. Overall, our results will help in understanding the immune molecular regulatory networks influenced by the ACE2 mediated interaction in other body tissues, which may aid in identifying the secondary health complications associated with SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Christy Lite
- Endocrine and Exposome (EE2) Laboratory, Madras Christian College, Department of Zoology, Tamil Nadu - 600059, India
| | - Shiek S S J Ahmed
- Drug Discovery Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu - 603103, India
| | - Melita Juliet
- Department of Oral and Maxillofacial Surgery, Meenakshi Ammal Dental College and Hospital, Maduravoyal, Chennai, Tamil Nadu - 600095, India
| | - A J Freddy
- Endocrine and Exposome (EE2) Laboratory, Madras Christian College, Department of Zoology, Tamil Nadu - 600059, India
| |
Collapse
|
47
|
Thankam FG, Agrawal DK. Molecular chronicles of cytokine burst in patients with coronavirus disease 2019 (COVID-19) with cardiovascular diseases. J Thorac Cardiovasc Surg 2021; 161:e217-e226. [PMID: 32631657 PMCID: PMC7834736 DOI: 10.1016/j.jtcvs.2020.05.083] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/12/2020] [Accepted: 05/16/2020] [Indexed: 02/02/2023]
Affiliation(s)
| | - Devendra K. Agrawal
- Address for reprints: Devendra K. Agrawal, PhD (Biochem), PhD (Med Sci), MBA, Department of Translational Research, Western University of Health Sciences, 309 E Second St, Pomona, CA 91766
| |
Collapse
|
48
|
ACE-2-interacting Domain of SARS-CoV-2 (AIDS) Peptide Suppresses Inflammation to Reduce Fever and Protect Lungs and Heart in Mice: Implications for COVID-19 Therapy. J Neuroimmune Pharmacol 2021; 16:59-70. [PMID: 33426604 PMCID: PMC7797355 DOI: 10.1007/s11481-020-09979-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 12/18/2020] [Indexed: 11/28/2022]
Abstract
COVID-19 is an infectious respiratory illness caused by the virus strain severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and until now, there is no effective therapy against COVID-19. Since SARS-CoV-2 binds to angiotensin-converting enzyme 2 (ACE2) for entering into host cells, to target COVID-19 from therapeutic angle, we engineered a hexapeptide corresponding to the ACE2-interacting domain of SARS-CoV-2 (AIDS) that inhibits the association between receptor-binding domain-containing spike S1 and ACE-2. Accordingly, wild type (wt), but not mutated (m), AIDS peptide inhibited SARS-CoV-2 spike S1-induced activation of NF-κB and expression of IL-6 in human lungs cells. Interestingly, intranasal intoxication of C57/BL6 mice with recombinant SARS-CoV-2 spike S1 led to fever, increase in IL-6 in lungs, infiltration of neutrophils into the lungs, arrhythmias, and impairment in locomotor activities, mimicking some of the important symptoms of COVID-19. However, intranasal treatment with wtAIDS, but not mAIDS, peptide reduced fever, protected lungs, improved heart function, and enhanced locomotor activities in SARS-CoV-2 spike S1-intoxicated mice. Therefore, selective targeting of ACE2-to-SARS-CoV-2 interaction by wtAIDS may be beneficial for COVID-19.
Collapse
|
49
|
Hammoud SH, Wehbe Z, Abdelhady S, Kobeissy F, Eid AH, El-Yazbi AF. Dysregulation of Angiotensin Converting Enzyme 2 Expression and Function in Comorbid Disease Conditions Possibly Contributes to Coronavirus Infectious Disease 2019 Complication Severity. Mol Pharmacol 2021; 99:17-28. [PMID: 33082267 DOI: 10.1124/molpharm.120.000119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/09/2020] [Indexed: 01/08/2023] Open
Abstract
ACE2 has emerged as a double agent in the COVID-19 ordeal, as it is both physiologically protective and virally conducive. The identification of ACE2 in as many as 72 tissues suggests that extrapulmonary invasion and damage is likely, which indeed has already been demonstrated by cardiovascular and gastrointestinal symptoms. On the other hand, identifying ACE2 dysregulation in patients with comorbidities may offer insight as to why COVID-19 symptoms are often more severe in these individuals. This may be attributed to a pre-existing proinflammatory state that is further propelled with the cytokine storm induced by SARS-CoV-2 infection or the loss of functional ACE2 expression as a result of viral internalization. Here, we aim to characterize the distribution and role of ACE2 in various organs to highlight the scope of damage that may arise upon SARS-CoV-2 invasion. Furthermore, by examining the disruption of ACE2 in several comorbid diseases, we offer insight into potential causes of increased severity of COVID-19 symptoms in certain individuals. SIGNIFICANCE STATEMENT: Cell surface expression of ACE2 determines the tissue susceptibility for coronavirus infectious disease 2019 infection. Comorbid disease conditions altering ACE2 expression could increase the patient's vulnerability for the disease and its complications, either directly, through modulation of viral infection, or indirectly, through alteration of inflammatory status.
Collapse
Affiliation(s)
- Safaa H Hammoud
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Zena Wehbe
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Samar Abdelhady
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Firas Kobeissy
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Ali H Eid
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| |
Collapse
|
50
|
Abstract
While clinical characteristics exhibit that susceptibility to COVID-19 infection is equally likely between males and females, clinical outcomes show that males experience both a higher severity and fatality for COVID-19 infection than females. This review examines the evidence for these sex and gender differences and aims to illustrate possible mechanisms behind such sensitivity. Successful entry of SARS-CoV-2 into the body is dependent on the angiotensin-converting enzyme 2 (ACE2) receptor and the transmembrane protease serine 2 (TMPRSS2). Thus, sex-based differences in the expression of the ACE2 receptor and TMPRSS2 may explain the disparities in COVID-19 severity and fatality. Furthermore, these disparities may also be attributed to sex-based difference in immunological responses. Finally, the differences in clinical outcomes of COVID-19 infections between men and women may be due to gendered differences in behaviors, such as smoking, and prevalence to comorbidities. An understanding of the sex and gender sensitivities of COVID-19 infection is a necessary component towards the creation of effective treatment options and therapies for the virus. Graphical abstract.
Collapse
Affiliation(s)
- Shreya Mukherjee
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St Suite Cohn 310, Chicago, IL 60612 USA ,Present Address: Honors Program in Medical Education, Northwestern University, Chicago, IL USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St Suite Cohn 310, Chicago, IL 60612 USA ,Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL USA
| |
Collapse
|